Journal articles on the topic 'Potassium channel'

To see the other types of publications on this topic, follow the link: Potassium channel.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Potassium channel.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Hopkins, W. F., J. L. Miller, and G. P. Miljanich. "Voltage-gated Potassium Channel Inhibitors." Current Pharmaceutical Design 2, no. 4 (August 1996): 389–96. http://dx.doi.org/10.2174/1381612802666220925203618.

Full text
Abstract:
Forty years have transpired since tetraethylammonium was first used to selectively inhibit the potassiuin conductance in squid axons. Since then, a large body of work has emerged describing inhibitors of voltage-gated potassium currents in a variety of cells. The advent of molecular cloning techniques and the cloning of the potassium channel encoded by the Shaker locus in Drosophila has enabled detailed structure­ function studies of several potassium channel subunits. These breakthroughs have also recently enabled studies of the "toxinology" and pharmacology of specific potassium channel subunits expressed heterologously in Xenopus oocytes and other cells. Here we describe the results of some of those efforts, focusing in particular on our work with four members of the Shaker subfamily of potassium channel a-subunits: Kvl.1 through Kvl.4. These subunits are expressed in the central nervous system and other tissues of rodents, and are highly homologous to corresponding subunits expressed in humans. We provide a profile of potency and selectivity for.five snake dendrotoxins as well as several scorpion toxins for these potassium channel subunits expressed in Xenopus oocytes. We also provide similar data for four other peptide toxins and several nonpeptide compounds that had previously been shown to inhibit potassium currents. We discuss several potential clinical applications of potassium channel inhibitors, including demyelinating diseases such as multiple sclerosis, immunosuppression, cardiac arrhythmias, neurodegenerative and psychiatric diseases. Further progress will require, among other things, a greater understanding of the expression patterns of potassium channel subunits in the CNS and elsewhere as well as knowledge of the specific subunit composition of heteromultimeric channels.
APA, Harvard, Vancouver, ISO, and other styles
2

Wrzosek, Antoni, Bartłomiej Augustynek, Monika Żochowska, and Adam Szewczyk. "Mitochondrial Potassium Channels as Druggable Targets." Biomolecules 10, no. 8 (August 18, 2020): 1200. http://dx.doi.org/10.3390/biom10081200.

Full text
Abstract:
Mitochondrial potassium channels have been described as important factors in cell pro-life and death phenomena. The activation of mitochondrial potassium channels, such as ATP-regulated or calcium-activated large conductance potassium channels, may have cytoprotective effects in cardiac or neuronal tissue. It has also been shown that inhibition of the mitochondrial Kv1.3 channel may lead to cancer cell death. Hence, in this paper, we examine the concept of the druggability of mitochondrial potassium channels. To what extent are mitochondrial potassium channels an important, novel, and promising drug target in various organs and tissues? The druggability of mitochondrial potassium channels will be discussed within the context of channel molecular identity, the specificity of potassium channel openers and inhibitors, and the unique regulatory properties of mitochondrial potassium channels. Future prospects of the druggability concept of mitochondrial potassium channels will be evaluated in this paper.
APA, Harvard, Vancouver, ISO, and other styles
3

Gomez-Sanchez, Celso E., and Kenji Oki. "Minireview: Potassium Channels and Aldosterone Dysregulation: Is Primary Aldosteronism a Potassium Channelopathy?" Endocrinology 155, no. 1 (January 1, 2014): 47–55. http://dx.doi.org/10.1210/en.2013-1733.

Full text
Abstract:
Primary aldosteronism is the most common form of secondary hypertension and has significant cardiovascular consequences. Aldosterone-producing adenomas (APAs) are responsible for half the cases of primary aldosteronism, and about half have mutations of the G protein-activated inward rectifying potassium channel Kir3.4. Under basal conditions, the adrenal zona glomerulosa cells are hyperpolarized with negative resting potentials determined by membrane permeability to K+ mediated through various K+ channels, including the leak K+ channels TASK-1, TASK-3, and Twik-Related Potassium Channel 1, and G protein inward rectifying potassium channel Kir3.4. Angiotensin II decreases the activity of the leak K+ channels and Kir3.4 channel and decreases the expression of the Kir3.4 channel, resulting in membrane depolarization, increased intracellular calcium, calcium-calmodulin pathway activation, and increased expression of cytochrome P450 aldosterone synthase (CYP11B2), the last enzyme for aldosterone production. Somatic mutations of the selectivity filter of the Kir3.4 channel in APA results in loss of selectivity for K+ and entry of sodium, resulting in membrane depolarization, calcium mobilization, increased CYP11B2 expression, and hyperaldosteronism. Germ cell mutations cause familial hyperaldosteronism type 3, which is associated with adrenal zona glomerulosa hyperplasia, rather than adenoma. Less commonly, somatic mutations of the sodium-potassium ATPase, calcium ATPase, or the calcium channel calcium channel voltage-dependent L type alpha 1D have been found in some APAs. The regulation of aldosterone secretion is exerted to a significant degree by activation of membrane K+ and calcium channels or pumps, so it is not surprising that the known causes of disorders of aldosterone secretion in APA have been channelopathies, which activate mechanisms that increase aldosterone synthesis.
APA, Harvard, Vancouver, ISO, and other styles
4

Huang, Xi, and Lily Yeh Jan. "Targeting potassium channels in cancer." Journal of Cell Biology 206, no. 2 (July 21, 2014): 151–62. http://dx.doi.org/10.1083/jcb.201404136.

Full text
Abstract:
Potassium channels are pore-forming transmembrane proteins that regulate a multitude of biological processes by controlling potassium flow across cell membranes. Aberrant potassium channel functions contribute to diseases such as epilepsy, cardiac arrhythmia, and neuromuscular symptoms collectively known as channelopathies. Increasing evidence suggests that cancer constitutes another category of channelopathies associated with dysregulated channel expression. Indeed, potassium channel–modulating agents have demonstrated antitumor efficacy. Potassium channels regulate cancer cell behaviors such as proliferation and migration through both canonical ion permeation–dependent and noncanonical ion permeation–independent functions. Given their cell surface localization and well-known pharmacology, pharmacological strategies to target potassium channel could prove to be promising cancer therapeutics.
APA, Harvard, Vancouver, ISO, and other styles
5

Vyas, Vivek K., Palak Parikh, Jonali Ramani, and Manjunath Ghate. "Medicinal Chemistry of Potassium Channel Modulators: An Update of Recent Progress (2011-2017)." Current Medicinal Chemistry 26, no. 12 (July 1, 2019): 2062–84. http://dx.doi.org/10.2174/0929867325666180430152023.

Full text
Abstract:
Background: Potassium (K+) channels participate in many physiological processes, cardiac function, cell proliferation, neuronal signaling, muscle contractility, immune function, hormone secretion, osmotic pressure, changes in gene expression, and are involved in critical biological functions, and in a variety of diseases. Potassium channels represent a large family of tetrameric membrane proteins. Potassium channels activation reduces excitability, whereas channel inhibition increases excitability. Objective: Small molecule K+ channel activators and inhibitors interact with voltage-gated, inward rectifying, and two-pore tandem potassium channels. Due to their involvement in biological functions, and in a variety of diseases, small molecules as potassium channel modulators have received great scientific attention. Methods: : In this review, we have compiled the literature, patents and patent applications (2011 to 2017) related to different chemical classes of potassium channel openers and blockers as therapeutic agents for the treatment of various diseases. Many different chemical classes of selective small molecule have emerged as potassium channel modulators over the past years. Conclusion: This review discussed the current understanding of medicinal chemistry research in the field of potassium channel modulators to update the key advances in this field.
APA, Harvard, Vancouver, ISO, and other styles
6

Zhao, Yonghui, Zongyun Chen, Zhijian Cao, Wenxin Li, and Yingliang Wu. "Diverse Structural Features of Potassium Channels Characterized by Scorpion Toxins as Molecular Probes." Molecules 24, no. 11 (May 29, 2019): 2045. http://dx.doi.org/10.3390/molecules24112045.

Full text
Abstract:
Scorpion toxins are well-known as the largest potassium channel peptide blocker family. They have been successfully proven to be valuable molecular probes for structural research on diverse potassium channels. The potassium channel pore region, including the turret and filter regions, is the binding interface for scorpion toxins, and structural features from different potassium channels have been identified using different scorpion toxins. According to the spatial orientation of channel turrets with differential sequence lengths and identities, conformational changes and molecular surface properties, the potassium channel turrets can be divided into the following three states: open state with less hindering effects on toxin binding, half-open state or half-closed state with certain effects on toxin binding, and closed state with remarkable effects on toxin binding. In this review, we summarized the diverse structural features of potassium channels explored using scorpion toxin tools and discuss future work in the field of scorpion toxin-potassium channel interactions.
APA, Harvard, Vancouver, ISO, and other styles
7

Spires, S., and T. Begenisich. "Modification of potassium channel kinetics by histidine-specific reagents." Journal of General Physiology 96, no. 4 (October 1, 1990): 757–75. http://dx.doi.org/10.1085/jgp.96.4.757.

Full text
Abstract:
We have examined the actions of histidine-specific reagents on potassium channels in squid giant axons. External application of 20-500 microM diethylpyrocarbonate (DEP) slowed the opening of potassium channels with little or no effect on closing rates. Sodium channels were not affected by these low external concentrations of DEP. Internal application of up to 2 mM DEP had no effect on potassium channel kinetics. Steady-state potassium channel currents were reduced in an apparently voltage-dependent manner by external treatment with this reagent. The shape of the instantaneous current-voltage relation was not altered. The voltage-dependent probability of channel opening was shifted toward more positive membrane potentials, thus accounting for the apparent voltage-dependent reduction of steady-state current. Histidine-specific photo-oxidation catalyzed by rose bengal produced alterations in potassium channel properties similar to those observed with DEP. The rate of action of DEP was consistent with a single kinetic class of histidine residues. In contrast to the effects on ionic currents, potassium channel gating currents were not modified by treatment with DEP. These results suggest the existence of a histidyl group (or groups) on the external surface of potassium channels important for a weakly voltage-dependent conformational transition. These effects can be reproduced by a simple kinetic model of potassium channels.
APA, Harvard, Vancouver, ISO, and other styles
8

Barman, Scott A. "Potassium channels modulate hypoxic pulmonary vasoconstriction." American Journal of Physiology-Lung Cellular and Molecular Physiology 275, no. 1 (July 1, 1998): L64—L70. http://dx.doi.org/10.1152/ajplung.1998.275.1.l64.

Full text
Abstract:
The role of Ca2+-activated K+-channel, ATP-sensitive K+-channel, and delayed rectifier K+-channel modulation in the canine pulmonary vascular response to hypoxia was determined in the isolated blood-perfused dog lung. Pulmonary vascular resistances and compliances were measured with vascular occlusion techniques. Under normoxia, the Ca2+-activated K+-channel blocker tetraethylammonium (1 mM), the ATP-sensitive K+-channel inhibitor glibenclamide (10−5 M), and the delayed rectifier K+-channel blocker 4-aminopyridine (10−4 M) elicited a small but significant increase in pulmonary arterial pressure. Hypoxia significantly increased pulmonary arterial and venous resistances and pulmonary capillary pressure and decreased total vascular compliance by decreasing both microvascular and large-vessel compliances. Tetraethylammonium, glibenclamide, and 4-aminopyridine potentiated the response to hypoxia on the arterial segments but not on the venous segments and also further decreased pulmonary vascular compliance. In contrast, the ATP-sensitive K+-channel opener cromakalim and the L-type voltage-dependent Ca2+-channel blocker verapamil (10−5 M) inhibited the vasoconstrictor effect of hypoxia on both the arterial and venous vessels. These results indicate that closure of the Ca2+-activated K+ channels, ATP-sensitive K+ channels, and delayed rectifier K+ channels potentiate the canine pulmonary arterial response under hypoxic conditions and that L-type voltage-dependent Ca2+ channels modulate hypoxic vasoconstriction. Therefore, the possibility exists that K+-channel inhibition is a key event that links hypoxia to pulmonary vasoconstriction by eliciting membrane depolarization and subsequent Ca2+-channel activation, leading to Ca2+ influx.
APA, Harvard, Vancouver, ISO, and other styles
9

Sun, Yuan-Kun, Liang-Hao Guo, Kai-Cheng Wang, Shao-Meng Wang, and Yu-Bin Gong. "Molecular dynamics simulation of effect of terahertz waves on the secondary structure of potassium channel proteins." Acta Physica Sinica 70, no. 24 (2021): 248701. http://dx.doi.org/10.7498/aps.70.20211725.

Full text
Abstract:
Potassium channels play an important role in repolarizing the nerve cell action potentials. There are many types of potassium channel proteins, and potassium channels allow potassium ions to specifically pass through the cell membrane, thereby maintaining the resting potential of nerve cells. In this paper, molecular dynamics simulation method is used to simulate the effects of 53.7 THz terahertz wave with different amplitudes on the secondary structure of KcsA potassium channel protein and the potassium ions rate. It is found in this study that under the action of the 53.7 THz terahertz wave, the number of alpha helices in KcsA potassium channel protein decreases, and the number of beta sheets and the number of coils increase. In addition, the 53.7 THz terahertz wave can accelerate potassium ions through the KcsA potassium channel. In this article, the effects of terahertz waves on potassium channel proteins are analyzed through the secondary structure of proteins, and a new perspective for the interaction between terahertz waves and biological functional molecules is presented as well.
APA, Harvard, Vancouver, ISO, and other styles
10

Park, Kang-Sik, Jae-Won Yang, Edward Seikel, and James S. Trimmer. "Potassium Channel Phosphorylation in Excitable Cells: Providing Dynamic Functional Variability to a Diverse Family of Ion Channels." Physiology 23, no. 1 (February 2008): 49–57. http://dx.doi.org/10.1152/physiol.00031.2007.

Full text
Abstract:
Phosphorylation of potassium channels affects their function and plays a major role in regulating cell physiology. Here, we review previous studies of potassium channel phosphorylation, focusing first on studies employing site-directed mutagenesis of recombinant channels expressed in heterologous cells. We then discuss recent mass spectrometric-based approaches to identify and quantify phosphorylation at specific sites on native and recombinant potassium channels, and newly developed mass spectrometric-based techniques that may prove beneficial to future studies of potassium channel phosphorylation, its regulation, and its mechanism of channel modulation.
APA, Harvard, Vancouver, ISO, and other styles
11

Kicinska, Anna, Bartlomiej Augustynek, Bogusz Kulawiak, Wieslawa Jarmuszkiewicz, Adam Szewczyk, and Piotr Bednarczyk. "A large-conductance calcium-regulated K+ channel in human dermal fibroblast mitochondria." Biochemical Journal 473, no. 23 (November 25, 2016): 4457–71. http://dx.doi.org/10.1042/bcj20160732.

Full text
Abstract:
Potassium channels have been found in the inner mitochondrial membrane of various cells. These channels regulate the mitochondrial membrane potential, respiration and production of reactive oxygen species. In the present study, we identified the activity of a mitochondrial large-conductance Ca2+-regulated potassium channel (mitoBKCa channel) in mitoplasts isolated from a primary human dermal fibroblast cell line. A potassium selective current was recorded with a mean conductance of 280 ± 2 pS in a symmetrical 150 mM KCl solution. The mitoBKCa channel was activated by the Ca2+ and by potassium channel opener NS1619. The channel activity was irreversibly inhibited by paxilline, a selective inhibitor of the BKCa channels. In isolated fibroblast mitochondria NS1619 depolarized the mitochondrial membrane potential, stimulated nonphosphorylating respiration and decreased superoxide formation. Additionally, the α- and β-subunits (predominantly the β3-form) of the BKCa channels were identified in fibroblast mitochondria. Our findings indicate, for the first time, the presence of a large-conductance Ca2+-regulated potassium channel in the inner mitochondrial membrane of human dermal fibroblasts.
APA, Harvard, Vancouver, ISO, and other styles
12

Abbott, Geoffrey W. "Biology of the KCNQ1 Potassium Channel." New Journal of Science 2014 (January 29, 2014): 1–26. http://dx.doi.org/10.1155/2014/237431.

Full text
Abstract:
Ion channels are essential for basic cellular function and for processes including sensory perception and intercellular communication in multicellular organisms. Voltage-gated potassium (Kv) channels facilitate dynamic cellular repolarization during an action potential, opening in response to membrane depolarization to facilitate K+ efflux. In both excitable and nonexcitable cells other, constitutively active, K+ channels provide a relatively constant repolarizing force to control membrane potential, ion homeostasis, and secretory processes. Of the forty known human Kv channel pore-forming α subunits that coassemble in various combinations to form the fundamental tetrameric channel pore and voltage sensor module, KCNQ1 is unique. KCNQ1 stands alone in having the capacity to form either channels that are voltage-dependent and require membrane depolarization for activation, or constitutively active channels. In mammals, KCNQ1 regulates processes including gastric acid secretion, thyroid hormone biosynthesis, salt and glucose homeostasis, and cell volume and in some species is required for rhythmic beating of the heart. In this review, the author discusses the unique functional properties, regulation, cell biology, diverse physiological roles, and involvement in human disease states of this chameleonic K+ channel.
APA, Harvard, Vancouver, ISO, and other styles
13

Ke, Wentao, Xiangdi Yu, and Yutong Gao. "Neonatal exposure to sevoflurane caused learning and memory impairment via dysregulating SK2 channel endocytosis." Science Progress 104, no. 3 (July 2021): 003685042110437. http://dx.doi.org/10.1177/00368504211043763.

Full text
Abstract:
Numerous studies have demonstrated that anesthetics’ exposure to neonates imposes toxicity on the developing brain but the underlying mechanisms need to be further elucidated. Our present study aimed to explore the role of small conductance Ca2+-activated potassium channel type2 in memory and learning dysfunction caused by exposing neonates to sevoflurane. Postnatal day 7 Sprague-Dawley rats and hemagglutinin-tagged small conductance Ca2+-activated potassium channel type2 channel transfected COS-7 cells were exposed to sevoflurane and the trafficking of small conductance Ca2+-activated potassium channel type2 channels was analyzed; furthermore, memory and learning ability was analyzed by the Morris water maze test on postnatal day30–35 (juvenile period). Our results showed that sevoflurane exposure inhibited small conductance Ca2+-activated potassium channel type2 channel endocytosis in both hippocampi of postnatal day 7 rats and hemagglutinin-tagged small conductance Ca2+-activated potassium channel type2 channel transfected COS-7 cells and the memory and learning ability was impaired in the juvenile period after sevoflurane exposure to neonatal rats. Herein, our results demonstrated that exposing neonates to sevoflurane caused memory and learning impairment via dysregulating small conductance Ca2+-activated potassium channel type2 channels endocytosis.
APA, Harvard, Vancouver, ISO, and other styles
14

Ogielska, Eva M., and Richard W. Aldrich. "A Mutation in S6 of Shaker Potassium Channels Decreases the K+ Affinity of an Ion Binding Site Revealing Ion–Ion Interactions in the Pore." Journal of General Physiology 112, no. 2 (August 1, 1998): 243–57. http://dx.doi.org/10.1085/jgp.112.2.243.

Full text
Abstract:
Under physiological conditions, potassium channels are extraordinarily selective for potassium over other ions. However, in the absence of potassium, certain potassium channels can conduct sodium. Sodium flux is blocked by the addition of low concentrations of potassium. Potassium affinity, and therefore the ability to block sodium current, varies among potassium channel subtypes (Korn, S.J., and S.R. Ikeda. 1995. Science. 269:410–412; Starkus, J.G., L. Kuschel, M.D. Rayner, and S.H. Heinemann. 1997. J. Gen. Physiol. 110:539–550). The Shaker potassium channel conducts sodium poorly in the presence of very low (micromolar) potassium due to its high potassium affinity (Starkus, J.G., L. Kuschel, M.D. Rayner, and S.H. Heinemann. 1997. J. Gen. Physiol. 110:539–550; Ogielska, E.M., and R.W. Aldrich. 1997. Biophys. J. 72:A233 [Abstr.]). We show that changing a single residue in S6, A463C, decreases the apparent internal potassium affinity of the Shaker channel pore from the micromolar to the millimolar range, as determined from the ability of potassium to block the sodium currents. Independent evidence that A463C decreases the apparent affinity of a binding site in the pore comes from a study of barium block of potassium currents. The A463C mutation decreases the internal barium affinity of the channel, as expected if barium blocks current by binding to a potassium site in the pore. The decrease in the apparent potassium affinity in A463C channels allows further study of possible ion interactions in the pore. Our results indicate that sodium and potassium can occupy the pore simultaneously and that multiple occupancy results in interactions between ions in the channel pore.
APA, Harvard, Vancouver, ISO, and other styles
15

HAMILTON, C. A., G. BERG, K. MCARTHUR, J. L. REID, and A. F. DOMINICZAK. "Does potassium channel opening contribute to endothelium-dependent relaxation in human internal thoracic artery?" Clinical Science 96, no. 6 (May 11, 1999): 631–38. http://dx.doi.org/10.1042/cs0960631.

Full text
Abstract:
Opening of potassium channels can cause hyperpolarization and relaxation of vascular smooth muscle cells. The aim of this work was to investigate the contribution of potassium channel activation to vasorelaxation in internal thoracic artery taken from patients undergoing coronary artery bypass graft surgery. Relaxations to carbachol and sodium nitroprusside were studied in isolated rings of internal thoracic artery in the absence and presence of nitric oxide synthase inhibitors and potassium channel blockers. The nitric oxide synthase inhibitors Nω-nitro-⌊-arginine methyl ester and NG-monomethyl-⌊-arginine abolished relaxations to carbachol. Relaxations to both carbachol and sodium nitroprusside were attenuated in the presence of raised extracellular potassium and the potassium channel blockers charybdotoxin, iberiotoxin and tetraethylammonium. Neither apamin nor glibenclamide modified relaxation. ODQ (1H-[1,2,4]oxadiazolol-[4,3a] quinoxalin-1-one), an inhibitor of soluble guanylate cyclase, abolished relaxation to carbachol in rings from some but not all subjects. These results suggest that potassium channel opening may make a small contribution to endothelium-dependent vasorelaxation in internal thoracic artery. The potassium channels had characteristics consistent with those of large-conductance calcium-dependent potassium channels.
APA, Harvard, Vancouver, ISO, and other styles
16

Liu, Renyu, Mayumi Ueda, Naoto Okazaki, and Yuichi Ishibe. "Role of Potassium Channels in Isoflurane- and Sevoflurane-induced Attenuation of Hypoxic Pulmonary Vasoconstriction in Isolated Perfused Rabbit Lungs." Anesthesiology 95, no. 4 (October 1, 2001): 939–46. http://dx.doi.org/10.1097/00000542-200110000-00024.

Full text
Abstract:
Background Although potassium channels are thought to be responsible for the initiation of hypoxic pulmonary vasoconstriction (HPV), their role in the HPV-inhibitory effect of volatile anesthetics is unclear. The current study tested if the HPV-inhibitory effect of isoflurane and sevoflurane can be affected by changing the potassium-channel opening status with specific potassium-channel inhibitors in isolated rabbit lungs. Methods Isolated rabbit lungs were divided into eight groups (n = 6 each in isoflurane groups and n = 8 in sevoflurane groups): those receiving no inhibitor treatment = control-isoflurane and control-sevoflurane groups; those treated with an adenosine triphosphate-sensitive potassium (K(ATP))-channel inhibitor, glibenclamide = glibenclamide-isoflurane and glibenclamide-sevoflurane groups; those treated with a high-conductance calcium-activated potassium (K(Ca))-channel inhibitor, iberiotoxin = iberiotoxin-isoflurane and iberiotoxin-sevoflurane groups; and those treated with a voltage-sensitive potassium (Kv)-channel inhibitor, 4-aminopyridine = 4-aminopyridine-isoflurane and 4-aminopyridine-sevoflurane groups. The effect of anesthetic on HPV was tested by exposure of the lungs to isoflurane at a concentration of 0, 0.5, 1, or 2 minimum alveolar concentration, or to sevoflurane at a concentration of 0, 0.5, 1, or 1.62 minimum alveolar concentration. The relation between anesthetic concentrations and the HPV response was analyzed by the Wagner equation. Results The inhibition of Kv channels by 4-aminopyridine and K(Ca) channels by iberiotoxin augmented the HPV response. The isoflurane-induced attenuation of HPV was attenuated by voltage-sensitive potassium-channel inhibition with 4-aminopyridine, potentiated by K(Ca)-channel inhibition with iberiotoxin, but not affected by K(ATP)-channel inhibition with glibenclamide. The sevoflurane-induced attenuation of HPV was not affected by any of the potassium-channel inhibitors. Conclusions Isoflurane may modulate the HPV response partially through K(Ca) and Kv channels, but sevoflurane may attenuate the HPV response through other pathways rather than through the currently investigated potassium channels in isolated rabbit lungs.
APA, Harvard, Vancouver, ISO, and other styles
17

Fox, J. A., B. A. Pfeffer, and G. L. Fain. "Single-channel recordings from cultured human retinal pigment epithelial cells." Journal of General Physiology 91, no. 2 (February 1, 1988): 193–222. http://dx.doi.org/10.1085/jgp.91.2.193.

Full text
Abstract:
We have applied patch-clamp techniques to on-cell and excised-membrane patches from human retinal pigment epithelial cells in tissue culture. Single-channel currents from at least four ion channel types were observed: three or more potassium-selective channels with single-channel slope conductances near 100, 45, and 25 pS as measured in on-cell patches with physiological saline in the pipette, and a relatively nonselective channel with subconductance states, which has a main-state conductance of approximately 300 pS at physiological ion concentrations. The permeability ratios, PK/PNa, measured in excised patches were 21 for the 100-pS channels, 3 for the 25-pS channels, and 0.8 for the 300-pS nonselective channel. The 45-pS channels appeared to be of at least two types, with PK/PNa's of approximately 41 for one type and 3 for the other. The potassium-selective channels were spontaneously active at all potentials examined. The average open time for these channels ranged from a few milliseconds to many tens of milliseconds. No consistent trend relating potassium-selective channel kinetics to membrane potential was apparent, which suggests that channel activity was not regulated by the membrane potential. In contrast to the potassium-selective channels, the activity of the nonselective channel was voltage dependent: the open probability of this channel declined to low values at large positive or negative membrane potentials and was maximal near zero. Single-channel conductances observed at several symmetrical KCl concentrations have been fitted with Michaelis-Menten curves in order to estimate maximum channel conductances and ion-binding constants for the different channel types. The channels we have recorded are probably responsible for the previously observed potassium permeability of the retinal pigment epithelium apical membrane.
APA, Harvard, Vancouver, ISO, and other styles
18

Perry, Matthew D., Vazhaikkurichi M. Rajendran, Kenneth A. MacLennan, and Geoffrey I. Sandle. "Segmental differences in upregulated apical potassium channels in mammalian colon during potassium adaptation." American Journal of Physiology-Gastrointestinal and Liver Physiology 311, no. 5 (November 1, 2016): G785—G793. http://dx.doi.org/10.1152/ajpgi.00181.2015.

Full text
Abstract:
Rat proximal and distal colon are net K+secretory and net K+absorptive epithelia, respectively. Chronic dietary K+loading increases net K+secretion in the proximal colon and transforms net K+absorption to net K+secretion in the distal colon, but changes in apical K+channel expression are unclear. We evaluated expression/activity of apical K+(BK) channels in surface colonocytes in proximal and distal colon of control and K+-loaded animals using patch-clamp recording, immunohistochemistry, and Western blot analyses. In controls, BK channels were more abundant in surface colonocytes from K+secretory proximal colon (39% of patches) than in those from K+-absorptive distal colon (12% of patches). Immunostaining demonstrated more pronounced BK channel α-subunit protein expression in surface cells and cells in the upper 25% of crypts in proximal colon, compared with distal colon. Dietary K+loading had no clear-cut effects on the abundance, immunolocalization, or expression of BK channels in proximal colon. By contrast, in distal colon, K+loading 1) increased BK channel abundance in patches from 12 to 41%; 2) increased density of immunostaining in surface cells, which extended along the upper 50% of crypts; and 3) increased expression of BK channel α-subunit protein when assessed by Western blotting ( P < 0.001). Thus apical BK channels are normally more abundant in K+secretory proximal colon than in K+absorptive distal colon, and apical BK channel expression in distal (but not proximal) colon is greatly stimulated as part of the enhanced K+secretory response to dietary K+loading.
APA, Harvard, Vancouver, ISO, and other styles
19

Yang, Youshan, and Fred J. Sigworth. "Single-Channel Properties of IKs Potassium Channels." Journal of General Physiology 112, no. 6 (December 1, 1998): 665–78. http://dx.doi.org/10.1085/jgp.112.6.665.

Full text
Abstract:
Expressed in Xenopus oocytes, KvLQT1 channel subunits yield a small, rapidly activating, voltage- dependent potassium conductance. When coexpressed with the minK gene product, a slowly activating and much larger potassium current results. Using fluctuation analysis and single-channel recordings, we have studied the currents formed by human KvLQT1 subunits alone and in conjunction with human or rat minK subunits. With low external K+, the single-channel conductances of these three channel types are estimated to be 0.7, 4.5, and 6.5 pS, respectively, based on noise analysis at 20 kHz bandwidth of currents at +50 mV. Power spectra computed over the range 0.1 Hz–20 kHz show a weak frequency dependence, consistent with current interruptions occurring on a broad range of time scales. The broad spectrum causes the apparent single-channel current value to depend on the bandwidth of the recording, and is mirrored in very “flickery” single-channel events of the channels from coexpressed KvLQT1 and human minK subunits. The increase in macroscopic current due to the presence of the minK subunit is accounted for by the increased apparent single-channel conductance it confers on the expressed channels. The rat minK subunit also confers the property that the outward single-channel current is increased by external potassium ions.
APA, Harvard, Vancouver, ISO, and other styles
20

Duty, Susan, and Arthur H. Weston. "Potassium Channel Openers." Drugs 40, no. 6 (December 1990): 785–91. http://dx.doi.org/10.2165/00003495-199040060-00002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Choe, Senyon. "Potassium channel structures." Nature Reviews Neuroscience 3, no. 2 (February 2002): 115–21. http://dx.doi.org/10.1038/nrn727.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Campbell, Jeff D., Mark S. P. Sansom, and Frances M. Ashcroft. "Potassium channel regulation." EMBO reports 4, no. 11 (October 2003): 1038–42. http://dx.doi.org/10.1038/sj.embor.7400003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Campbell, Jeff D., Mark S. P. Sansom, and Frances M. Ashcroft. "Potassium channel regulation." EMBO reports 4, no. 11 (November 2003): 1038–42. http://dx.doi.org/10.1038/sj.embor.embor7400003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

&NA;. "Potassium channel activators." Inpharma Weekly &NA;, no. 836 (May 1992): 9–10. http://dx.doi.org/10.2165/00128413-199208360-00014.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

REES, S. A. "Potassium Channel Modulators." Cardiovascular Research 27, no. 10 (October 1, 1993): 1887. http://dx.doi.org/10.1093/cvr/27.10.1887.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Sigworth, Fred J. "Potassium Channel Mechanics." Neuron 32, no. 4 (November 2001): 555–56. http://dx.doi.org/10.1016/s0896-6273(01)00509-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Deutsch, Carol. "Potassium Channel Ontogeny." Annual Review of Physiology 64, no. 1 (March 2002): 19–46. http://dx.doi.org/10.1146/annurev.physiol.64.081501.155934.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Strong, P. N. "Potassium channel toxins." Pharmacology & Therapeutics 46, no. 1 (January 1990): 137–62. http://dx.doi.org/10.1016/0163-7258(90)90040-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Cheng, Wayland W. L., Decha Enkvetchakul, and Colin G. Nichols. "KirBac1.1: It's an Inward Rectifying Potassium Channel." Journal of General Physiology 133, no. 3 (February 9, 2009): 295–305. http://dx.doi.org/10.1085/jgp.200810125.

Full text
Abstract:
KirBac1.1 is a prokaryotic homologue of eukaryotic inward rectifier potassium (Kir) channels. The crystal structure of KirBac1.1 and related KirBac3.1 have now been used extensively to generate in silico models of eukaryotic Kir channels, but functional analysis has been limited to 86Rb+ flux experiments and bacteria or yeast complementation screens, and no voltage clamp analysis has been available. We have expressed pure full-length His-tagged KirBac1.1 protein in Escherichia coli and obtained voltage clamp recordings of recombinant channel activity in excised membrane patches from giant liposomes. Macroscopic currents of wild-type KirBac1.1 are K+ selective and spermine insensitive, but blocked by Ba2+, similar to “weakly rectifying” eukaryotic Kir1.1 and Kir6.2 channels. The introduction of a negative charge at a pore-lining residue, I138D, generates high spermine sensitivity, similar to that resulting from the introduction of a negative charge at the equivalent position in Kir1.1 or Kir6.2. KirBac1.1 currents are also inhibited by PIP2, consistent with 86Rb+ flux experiments, and reversibly inhibited by short-chain di-c8-PIP2. At the single-channel level, KirBac1.1 channels show numerous conductance states with two predominant conductances (15 pS and 32 pS at −100 mV) and marked variability in gating kinetics, similar to the behavior of KcsA in recombinant liposomes. The successful patch clamping of KirBac1.1 confirms that this prokaryotic channel behaves as a bona fide Kir channel and opens the way for combined biochemical, structural, and electrophysiological analysis of a tractable model Kir channel, as has been successfully achieved for the archetypal K+ channel KcsA.
APA, Harvard, Vancouver, ISO, and other styles
30

Volk, K. A., and E. F. Shibata. "Single delayed rectifier potassium channels from rabbit coronary artery myocytes." American Journal of Physiology-Heart and Circulatory Physiology 264, no. 4 (April 1, 1993): H1146—H1153. http://dx.doi.org/10.1152/ajpheart.1993.264.4.h1146.

Full text
Abstract:
Cell-attached patches from rabbit coronary artery single smooth muscle cells contained two distinct potassium channel types, namely a large conductance calcium-activated potassium channel and a smaller voltage-activated potassium channel representing the delayed rectifier (IK). When a physiological potassium ion gradient was used, the average slope conductance of single IK channels was 7.26 pS. The time course of activation measured from ensemble averages was well fit by a single exponential raised to the power of 2 and was voltage dependent. Experiments were then performed with potassium (140 mM) on both sides of the membrane to resolve single IK channel currents during deactivation. Ensemble averages of this activity were well described by a two-component exponential, and the time constants were voltage dependent. Mean open times were significantly shorter during deactivation than during activation. Closed time distributions typically had two components. These kinetic characteristics were used in testing various state models for voltage-dependent potassium channels.
APA, Harvard, Vancouver, ISO, and other styles
31

Zorn, Lynda, Rama Kulkarni, Vellareddy Anantharam, Hagan Bayley, and Steven N. Treistman. "Halothane acts on many potassium channels, including a minimal potassium channel." Neuroscience Letters 161, no. 1 (October 1993): 81–84. http://dx.doi.org/10.1016/0304-3940(93)90145-b.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Sek, Aleksandra, Rafal P. Kampa, Bogusz Kulawiak, Adam Szewczyk, and Piotr Bednarczyk. "Identification of the Large-Conductance Ca2+-Regulated Potassium Channel in Mitochondria of Human Bronchial Epithelial Cells." Molecules 26, no. 11 (May 27, 2021): 3233. http://dx.doi.org/10.3390/molecules26113233.

Full text
Abstract:
Mitochondria play a key role in energy metabolism within the cell. Potassium channels such as ATP-sensitive, voltage-gated or large-conductance Ca2+-regulated channels have been described in the inner mitochondrial membrane. Several hypotheses have been proposed to describe the important roles of mitochondrial potassium channels in cell survival and death pathways. In the current study, we identified two populations of mitochondrial large-conductance Ca2+-regulated potassium (mitoBKCa) channels in human bronchial epithelial (HBE) cells. The biophysical properties of the channels were characterized using the patch-clamp technique. We observed the activity of the channel with a mean conductance close to 285 pS in symmetric 150/150 mM KCl solution. Channel activity was increased upon application of the potassium channel opener NS11021 in the micromolar concentration range. The channel activity was completely inhibited by 1 µM paxilline and 300 nM iberiotoxin, selective inhibitors of the BKCa channels. Based on calcium and iberiotoxin modulation, we suggest that the C-terminus of the protein is localized to the mitochondrial matrix. Additionally, using RT-PCR, we confirmed the presence of α pore-forming (Slo1) and auxiliary β3-β4 subunits of BKCa channel in HBE cells. Western blot analysis of cellular fractions confirmed the mitochondrial localization of α pore-forming and predominately β3 subunits. Additionally, the regulation of oxygen consumption and membrane potential of human bronchial epithelial mitochondria in the presence of the potassium channel opener NS11021 and inhibitor paxilline were also studied. In summary, for the first time, the electrophysiological and functional properties of the mitoBKCa channel in a bronchial epithelial cell line were described.
APA, Harvard, Vancouver, ISO, and other styles
33

Sackin, H., and L. G. Palmer. "Basolateral potassium channels in renal proximal tubule." American Journal of Physiology-Renal Physiology 253, no. 3 (September 1, 1987): F476—F487. http://dx.doi.org/10.1152/ajprenal.1987.253.3.f476.

Full text
Abstract:
Potassium (K+) channels in the basolateral membrane of unperfused Necturus proximal tubules were studied in both cell-attached and excised patches, after removal of the tubule basement membrane by manual dissection without collagenase. Two different K+ channels were identified on the basis of their kinetics: a short open-time K+ channel, with a mean open time less than 1 ms, and a long open-time K+ channel with a mean open time greater than 20 ms. The short open-time channel occurred more frequently than the longer channel, especially in excised patches. For inside-out excised patches with Cl- replaced by gluconate, the current-voltage relation of the short open-time K+ channel was linear over +/- 60 mV, with a K+-Na+ selectivity of 12 +/- 2 (n = 12), as calculated from the reversal potential with oppositely directed Na+ and K+ gradients. With K-Ringer in the patch pipette and Na-Ringer in the bath, the conductance of the short open-time channel was 47 +/- 2 pS (n = 15) for cell-attached patches, 26 +/- 2 pS (n = 15) for patches excised (inside out) into Na-Ringer, and 36 +/- 6 pS (n = 3) for excised patches with K-Ringer on both sides. These different conductances can be partially explained by a dependence of single-channel conductance on the K+ concentration on the interior side of the membrane. In experiments with a constant K+ gradient across excised patches, large changes in Na+ at the interior side of the membrane produced no change in single-channel conductance, arguing against a direct block of the K+ channel by Na+. Finally, the activity of the short open-time channel was voltage gated, where the mean number of open channels decreased as a linear function of basolateral membrane depolarization for potentials between -60 and 0 mV. Depolarization from -60 to -40 mV decreased the mean number of open K+ channels by 28 +/- 8% (n = 6).
APA, Harvard, Vancouver, ISO, and other styles
34

Mangel, A. W., V. Prpic, N. D. Snow, S. Basavappa, L. J. Hurst, A. I. Sharara, and R. A. Liddle. "Regulation of cholecystokinin secretion by ATP-sensitive potassium channels." American Journal of Physiology-Gastrointestinal and Liver Physiology 267, no. 4 (October 1, 1994): G595—G600. http://dx.doi.org/10.1152/ajpgi.1994.267.4.g595.

Full text
Abstract:
The relationship of potassium channel activity to the secretion of cholecystokinin (CCK) was evaluated in STC-1 cells, an intestinal CCK-secreting cell line. Patch-clamp and 86Rb efflux studies showed that an ATP-sensitive potassium channel was endogenously expressed in STC-1 cells. Furthermore, channels are present in sufficient number to significantly modulate whole cell potassium permeability after either channel activation or closure with diazoxide (100 microM) or disopyramide (200 microM), respectively. Inhibition of channel activity with glucose (5-20 mM) was found to depolarize the plasma membrane, increase cytosolic calcium levels, and stimulate CCK release. Glucose-mediated release of CCK, as well as the increase in cytosolic calcium, was inhibited by the calcium channel blocker diltiazem (10 microM). It is concluded that intestinal secretion of CCK may be tonically controlled by activity of basally active ATP-sensitive potassium channels, and after inhibition of channel activity, calcium-dependent CCK secretion is stimulated.
APA, Harvard, Vancouver, ISO, and other styles
35

Lawson, Kim. "Is there a Therapeutic Future for ‘Potassium Channel Openers’?" Clinical Science 91, no. 6 (December 1, 1996): 651–63. http://dx.doi.org/10.1042/cs0910651.

Full text
Abstract:
1. Potassium channels, which control cell electrical activity, are among the most regulated of all ion channels in biology. Promotion of activity in K+ channels by a wide range of physiological factors tends to stabilize cell function. 2. The discovery of synthetic molecules (e.g. cromakalim) that ‘directly’ open ATP-sensitive K+ channels has led to a new direction in pharmacology. ATP-sensitive K+ channel-opening properties have subsequently been demonstrated in a diverse range of chemical structures (synthetic and endogenous). 3. The existence of so many different subtypes of K+ channels has been an impetus in the search of new potassium channel openers with different channel selectivities and thus biological profiles. 4. The decrease in cell excitability following K+ channel opening implies a broad clinical potential in a number of pathological conditions for K+ channel openers. Preclinical and clinical evidence supports therapeutic roles of K+ channel openers in disorders of a wide range of biological cells. 5. Although lack of selectivity of current compounds remains a major hurdle, advances in K+ channel openers and K+ channel pharmacology are encouraging. Differences already observed in the pharmacology of K+ channel openers are important factors for the development of second-generation compounds, when tissue selectivity is sought. 6. The availability of subtype-selective K+ channel openers will facilitate detailed study, through a combined effort of electrophysiology, functional pharmacology and molecular biology, leading to focused therapeutic approaches for defined pathological conditions.
APA, Harvard, Vancouver, ISO, and other styles
36

Xia, Zhenglin, Xusen Huang, Kaiyun Chen, Hanning Wang, Jinfeng Xiao, Ke He, Rui Huang, et al. "Proapoptotic Role of Potassium Ions in Liver Cells." BioMed Research International 2016 (2016): 1–10. http://dx.doi.org/10.1155/2016/1729135.

Full text
Abstract:
Potassium channels are transmembrane proteins that selectively promote the infiltration of potassium ions. The significance of these channels for tumor biology has become obvious. However, the effects of potassium ions on the tumor or normal cells have seldom been studied. To address this problem, we studied the biological effects of L02 and HepG2 cells with ectogenous potassium ions. Cell proliferation, cell cycle, and apoptosis rate were analyzed. Our results indicated that potassium ions inhibited proliferation of L02 and HepG2 cells and promoted their apoptosis. Potassium ions induced apoptosis through regulating Bcl-2 family members and depolarized the mitochondrial membrane, especially for HepG2 cell. These biological effects were associated with channel protein HERG. By facilitating expression of channel protein HERG, potassium ions may prevent it from being shunted to procancerous pathways by inducing apoptosis. These results demonstrated that potassium ions may be a key regulator of liver cell function. Thus, our findings suggest that potassium ions could inhibit tumorigenesis through inducing apoptosis of hepatoma cells by upregulating potassium ions transport channel proteins HERG and VDAC1.
APA, Harvard, Vancouver, ISO, and other styles
37

Haris, Parvez I. "Synthetic Peptide Fragments as Probes for Structure Determination of Potassium Ion-Channel Proteins." Bioscience Reports 18, no. 6 (December 1, 1998): 299–312. http://dx.doi.org/10.1023/a:1020257215577.

Full text
Abstract:
Potassium channels are a diverse class of transmembrane proteins that are responsible for diffusion of potassium ion across cell membranes. The lack of large quantities of these proteins from natural sources, is a major hindrance in their structural characterization using biophysical techniques. Synthetic peptide fragments corresponding to functionally important domains of these proteins provide an attractive approach towards characterizing the structural organization of these ion-channels. Conformational properties of peptides from three different potassium channels (Shaker, ROMK1 and minK) have been characterized in aqueous media, organic solvents and in phospholipid membranes. Techniques used for these studies include FTIR, CD and 2D-NMR spectroscopy. FTIR spectroscopy has been a particularly valuable tool for characterizing the folding of the ion-channel peptides in phospholipid membranes; the three different types of potassium channels all share a common transmembrane folding pattern that is composed of a predominantly α-helical structure. There is no evidence to suggest the presence of any significant β-sheet structure. These results are in excellent agreement with the crystal structure of a bacterial potassium channel (Doyle, D. A. et al. (1998) Science280:69–77), and suggest that all potassium channel proteins may share a common folding motif where the ion-channel structure is constructed entirely from α-helices.
APA, Harvard, Vancouver, ISO, and other styles
38

Beacham, Daniel W., Trillium Blackmer, Michael O’ Grady, and George T. Hanson. "Cell-Based Potassium Ion Channel Screening Using the FluxOR™ Assay." Journal of Biomolecular Screening 15, no. 4 (March 5, 2010): 441–46. http://dx.doi.org/10.1177/1087057109359807.

Full text
Abstract:
FluxOR™ technology is a cell-based assay used for high-throughput screening measurements of potassium channel activity. Using thallium influx as a surrogate indicator of potassium ion channel activity, the FluxOR™ Potassium Ion Channel Assay is based on the activation of a novel fluorescent dye. This indicator reports channel activity with a large fluorogenic response and is proportional to the number of open potassium channels on the cell, making it extremely useful for studying K+ channel targets. In contrast to BTC-AM ester, FluxOR™ dye is roughly 10-fold more thallium sensitive, requiring much lower thallium for a larger signal window. This also means that the assay is carried out in a physiological, normal-chloride saline. In this article, the authors describe how they used BacMam gene delivery to express Kv7.2 and 7.3 (KCNQ), Kir2.1, or Kv11.1 (hERG) potassium ion channels in U2-OS cells. Using these cells, they ran the FluxOR™ assay to identify and characterize channel-specific inhibitory compounds discovered within the library (Tocriscreen™ Mini 1200 and Sigma Sodium/Potassium Modulators Ligand set). The FluxOR™ assay was able to identify several known specific inhibitors of Kv7.2/7.3 or hERG, highlighting its potential to identify novel and more efficacious small-molecule modulators.
APA, Harvard, Vancouver, ISO, and other styles
39

Khalid, Rida, Nighat Noureen, Mohammad Amjad Kamal, and Sidra Batool. "Computational Protein-Protein Docking Reveals the Therapeutic Potential of Kunitz-type Venom against hKv1.2 Binding Sites." CNS & Neurological Disorders - Drug Targets 18, no. 5 (September 23, 2019): 382–404. http://dx.doi.org/10.2174/1871527318666190319140204.

Full text
Abstract:
Background & Objective: Kunitz-type venoms are bioactive proteins isolated from a wide variety of venomous animals. These venoms are involved in protease inhibitory activity or potassium channel blocking activity. Therefore, they are reported as an important source for lead drug candidates towards protease or channel associated diseases like neurological, metabolic and cardiovascular disorders. Methods: This study aimed to check the inhibitory action of Kunitz-type venoms against potassium channels using computational tools. Results: Among potassium channels, Human Voltage-Gated Potassium Channel 1.2 (hKv1.2) was used as a receptor whereas Kunitz-type peptides from the venoms of various species were selected as ligand dataset. Conclusion: This study helped in finding the binding interface between the receptor and ligand dataset for their potential therapeutic use in treating potassium channelopathies.
APA, Harvard, Vancouver, ISO, and other styles
40

El Hachmane, Mickael-F., Kathryn A. Rees, Emma L. Veale, Vadim V. Sumbayev, and Alistair Mathie. "Enhancement of TWIK-related Acid-sensitive Potassium Channel 3 (TASK3) Two-pore Domain Potassium Channel Activity by Tumor Necrosis Factor α." Journal of Biological Chemistry 289, no. 3 (December 4, 2013): 1388–401. http://dx.doi.org/10.1074/jbc.m113.500033.

Full text
Abstract:
TASK3 two-pore domain potassium (K2P) channels are responsible for native leak K channels in many cell types which regulate cell resting membrane potential and excitability. In addition, TASK3 channels contribute to the regulation of cellular potassium homeostasis. Because TASK3 channels are important for cell viability, having putative roles in both neuronal apoptosis and oncogenesis, we sought to determine their behavior under inflammatory conditions by investigating the effect of TNFα on TASK3 channel current. TASK3 channels were expressed in tsA-201 cells, and the current through them was measured using whole cell voltage clamp recordings. We show that THP-1 human myeloid leukemia monocytes, co-cultured with hTASK3-transfected tsA-201 cells, can be activated by the specific Toll-like receptor 7/8 activator, R848, to release TNFα that subsequently enhances hTASK3 current. Both hTASK3 and mTASK3 channel activity is increased by incubation with recombinant TNFα (10 ng/ml for 2–15 h), but other K2P channels (hTASK1, hTASK2, hTREK1, and hTRESK) are unaffected. This enhancement by TNFα is not due to alterations in levels of channel expression at the membrane but rather to an alteration in channel gating. The enhancement by TNFα can be blocked by extracellular acidification but persists for mutated TASK3 (H98A) channels that are no longer acid-sensitive even in an acidic extracellular environment. TNFα action on TASK3 channels is mediated through the intracellular C terminus of the channel. Furthermore, it occurs through the ASK1 pathway and is JNK- and p38-dependent. In combination, TNFα activation and TASK3 channel activity can promote cellular apoptosis.
APA, Harvard, Vancouver, ISO, and other styles
41

Walewska, Agnieszka, Adam Szewczyk, and Piotr Koprowski. "Gas Signaling Molecules and Mitochondrial Potassium Channels." International Journal of Molecular Sciences 19, no. 10 (October 18, 2018): 3227. http://dx.doi.org/10.3390/ijms19103227.

Full text
Abstract:
Recently, gaseous signaling molecules, such as carbon monoxide (CO), nitric oxide (NO), and hydrogen sulfide (H2S), which were previously considered to be highly toxic, have been of increasing interest due to their beneficial effects at low concentrations. These so-called gasotransmitters affect many cellular processes, such as apoptosis, proliferation, cytoprotection, oxygen sensing, ATP synthesis, and cellular respiration. It is thought that mitochondria, specifically their respiratory complexes, constitute an important target for these gases. On the other hand, increasing evidence of a cytoprotective role for mitochondrial potassium channels provides motivation for the analysis of the role of gasotransmitters in the regulation of channel function. A number of potassium channels have been shown to exhibit activity within the inner mitochondrial membrane, including ATP-sensitive potassium channels, Ca2+-activated potassium channels, voltage-gated Kv potassium channels, and TWIK-related acid-sensitive K+ channel 3 (TASK-3). The effects of these channels include the regulation of mitochondrial respiration and membrane potential. Additionally, they may modulate the synthesis of reactive oxygen species within mitochondria. The opening of mitochondrial potassium channels is believed to induce cytoprotection, while channel inhibition may facilitate cell death. The molecular mechanisms underlying the action of gasotransmitters are complex. In this review, we focus on the molecular mechanisms underlying the action of H2S, NO, and CO on potassium channels present within mitochondria.
APA, Harvard, Vancouver, ISO, and other styles
42

Hamilton, K. L., and D. C. Eaton. "cAMP-induced potassium channel activity in apical membrane of cultured A6 kidney cells." American Journal of Physiology-Renal Physiology 261, no. 6 (December 1, 1991): F1055—F1062. http://dx.doi.org/10.1152/ajprenal.1991.261.6.f1055.

Full text
Abstract:
In the present study, we report an adenosine 3',5'-cyclic monophosphate (cAMP)-induced potassium channel in the apical membrane of cultured A6 kidney cells grown on impermeable supports. The channel is present in approximately 10% of untreated cell-attached patches. After treatment with 1 mM dibutyryl-cAMP, the channel is present in greater than 70% of the same patches. The characteristics of this channel are 1) the channel is highly selective for potassium; 2) the channel has a unit conductance of 13 +/- 2 pS; 3) the probability of a channel opening increases in the presence of membrane permeable analogues of cAMP and with increasing depolarization of the cell interior; 4) channels are blocked by Ba2+; 5) the channel loses activity rapidly in excised patches; and 6) the channel has at least one open and two closed states. The mean open time is 3.5 +/- 1.0 ms, whereas the mean durations of the closed states are 3.2 +/- 1.4 and 29.4 +/- 3.4 ms. The channels could mediate potassium secretion in A6 cells, if the channels are normally present under transporting conditions; however, surface expression of the channels appears to depend on growth substrate and the state of cellular differentiation, since the channels are not observed in cells grown on permeable supports.
APA, Harvard, Vancouver, ISO, and other styles
43

Nakamura, Kazuyoshi, Hikaru Hayashi, and Manabu Kubokawa. "Proinflammatory Cytokines and Potassium Channels in the Kidney." Mediators of Inflammation 2015 (2015): 1–8. http://dx.doi.org/10.1155/2015/362768.

Full text
Abstract:
Proinflammatory cytokines affect several cell functions via receptor-mediated processes. In the kidney, functions of transporters and ion channels along the nephron are also affected by some cytokines. Among these, alteration of activity of potassium ion (K+) channels induces changes in transepithelial transport of solutes and water in the kidney, since K+channels in tubule cells are indispensable for formation of membrane potential which serves as a driving force for the transepithelial transport. Altered K+channel activity may be involved in renal cell dysfunction during inflammation. Although little information was available regarding the effects of proinflammatory cytokines on renal K+channels, reports have emerged during the last decade. In human proximal tubule cells, interferon-γshowed a time-dependent biphasic effect on a 40 pS K+channel, that is, delayed suppression and acute stimulation, and interleukin-1βacutely suppressed the channel activity. Transforming growth factor-β1 activated KCa3.1 K+channel in immortalized human proximal tubule cells, which would be involved in the pathogenesis of renal fibrosis. This review discusses the effects of proinflammatory cytokines on renal K+channels and the causal relationship between the cytokine-induced changes in K+channel activity and renal dysfunction.
APA, Harvard, Vancouver, ISO, and other styles
44

Williams, Beatrice A., and Keith J. Buckler. "Biophysical properties and metabolic regulation of a TASK-like potassium channel in rat carotid body type 1 cells." American Journal of Physiology-Lung Cellular and Molecular Physiology 286, no. 1 (January 2004): L221—L230. http://dx.doi.org/10.1152/ajplung.00010.2003.

Full text
Abstract:
The single channel properties of TASK-like oxygen-sensitive potassium channels were studied in rat carotid body type 1 cells. We observed channels with rapid bursting kinetics, active at resting membrane potentials. These channels were highly potassium selective with a slope conductance of 14–16 pS, values similar to those reported for TASK-1. In the absence of extracellular divalent cations, however, single channel conductance increased to 28 pS in a manner similar to that reported for TASK-3. After patch excision, channel activity ran down rapidly. Channel activity in inside-out patches was markedly increased by 2 and 5 mM ATP and by 2 mM ADP but not by 100 μM ADP or 1 mM AMP. In cell-attached patches, both cyanide and 2,4-dinitrophenol strongly inhibited channel activity. We conclude that 1) whilst the properties of this channel are consistent with it being a TASK-like potassium channel they do not precisely conform to those of either TASK-1 or TASK-3, 2) channel activity is highly dependent on cytosolic factors including ATP, and 3) changes in energy metabolism may play a role in regulating the activity of these background K+ channels.
APA, Harvard, Vancouver, ISO, and other styles
45

Lee, Hung-Hao, Po-Chao Hsu, Tsung-Hsien Lin, Wen-Ter Lai, and Sheng-Hsiung Sheu. "Nicorandil-Induced Hyperkalemia in a Uremic Patient." Case Reports in Medicine 2012 (2012): 1–4. http://dx.doi.org/10.1155/2012/812178.

Full text
Abstract:
Nicorandil is an antianginal agent with nitrate-like and ATP-sensitive potassium channel activator properties. After activation of potassium channels, potassium ions are expelled out of the cells, which lead to membrane hyperpolarization, closure of voltage-gated calcium channels, and finally vasodilation. We present a uremic case suffering from repeated junctional bradycardia, especially before hemodialysis. After detailed evaluation, nicorandil was suspected to be the cause of hyperkalemia which induced bradycardia. This case reminds us that physicians should be aware of this potential complication in patients receiving ATP-sensitive potassium channel activator.
APA, Harvard, Vancouver, ISO, and other styles
46

Grigoriev, Nikita G., J. David Spafford, and Andrew N. Spencer. "Modulation of Jellyfish Potassium Channels by External Potassium Ions." Journal of Neurophysiology 82, no. 4 (October 1, 1999): 1728–39. http://dx.doi.org/10.1152/jn.1999.82.4.1728.

Full text
Abstract:
The amplitude of an A-like potassium current ( I Kfast) in identified cultured motor neurons isolated from the jellyfish Polyorchis penicillatus was found to be strongly modulated by extracellular potassium ([K+]out). When expressed in Xenopus oocytes, two jellyfish Shaker-like genes, jShak1 and jShak2, coding for potassium channels, exhibited similar modulation by [K+]out over a range of concentrations from 0 to 100 mM. jShak2-encoded channels also showed a decreased rate of inactivation and an increased rate of recovery from inactivation at high [K+]out. Using site-directed mutagenesis we show that inactivation of jShak2 can be ascribed to an unusual combination of a weak “implicit” N-type inactivation mechanism and a strong, fast, potassium-sensitive C-type mechanism. Interaction between the two forms of inactivation is responsible for the potassium dependence of cumulative inactivation. Inactivation of jShak1 was determined primarily by a strong “ball and chain” mechanism similar to fruit fly Shaker channels. Experiments using fast perfusion of outside-out patches with jShak2 channels were used to establish that the effects of [K+]out on the peak current amplitude and inactivation were due to processes occurring at either different sites located at the external channel mouth with different retention times for potassium ions, or at the same site(s) where retention time is determined by state-dependent conformations of the channel protein. The possible physiological implications of potassium sensitivity of high-threshold potassium A-like currents is discussed.
APA, Harvard, Vancouver, ISO, and other styles
47

Liin, Sara I., Per-Eric Lund, Johan E. Larsson, Johan Brask, Björn Wallner, and Fredrik Elinder. "Biaryl sulfonamide motifs up- or down-regulate ion channel activity by activating voltage sensors." Journal of General Physiology 150, no. 8 (July 12, 2018): 1215–30. http://dx.doi.org/10.1085/jgp.201711942.

Full text
Abstract:
Voltage-gated ion channels are key molecules for the generation of cellular electrical excitability. Many pharmaceutical drugs target these channels by blocking their ion-conducting pore, but in many cases, channel-opening compounds would be more beneficial. Here, to search for new channel-opening compounds, we screen 18,000 compounds with high-throughput patch-clamp technology and find several potassium-channel openers that share a distinct biaryl-sulfonamide motif. Our data suggest that the negatively charged variants of these compounds bind to the top of the voltage-sensor domain, between transmembrane segments 3 and 4, to open the channel. Although we show here that biaryl-sulfonamide compounds open a potassium channel, they have also been reported to block sodium and calcium channels. However, because they inactivate voltage-gated sodium channels by promoting activation of one voltage sensor, we suggest that, despite different effects on the channel gates, the biaryl-sulfonamide motif is a general ion-channel activator motif. Because these compounds block action potential–generating sodium and calcium channels and open an action potential–dampening potassium channel, they should have a high propensity to reduce excitability. This opens up the possibility to build new excitability-reducing pharmaceutical drugs from the biaryl-sulfonamide scaffold.
APA, Harvard, Vancouver, ISO, and other styles
48

Shehata, Mohamed, Wojciech Kopec, Bert L. de Groot, and Rommie E. Amaro. "The potassium-selective channel “NaK2K”: Why is it a potassium channel?" Biophysical Journal 122, no. 3 (February 2023): 245a. http://dx.doi.org/10.1016/j.bpj.2022.11.1429.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Lindauer, Ute, Johannes Vogt, Sigrid Schuh-Hofer, Jens P. Dreier, and Ulrich Dirnagl. "Cerebrovascular Vasodilation to Extraluminal Acidosis Occurs via Combined Activation of ATP-Sensitive and Ca2+-Activated Potassium Channels." Journal of Cerebral Blood Flow & Metabolism 23, no. 10 (October 2003): 1227–38. http://dx.doi.org/10.1097/01.wcb.0000088764.02615.b7.

Full text
Abstract:
Albeit controversely discussed, it has been suggested by several authors that nitric oxide (NO) serves as a permissive factor in the cerebral blood flow response to systemic hypercapnia. Potassium channels are important regulators of cerebrovascular tone and may be modulated by a basal perivascular NO level. To elucidate the functional targets of the proposed NO modulation during hypercapnia-induced vasodilation, the authors performed experiments in isolated, cannulated, and pressurized rat middle cerebral arteries (MCA). Extracellular pH was reduced from 7.4 to 7.0 in the extraluminal bath to induce NO dependent vasdilation. Acidosis increased vessel diameter by 35 ± 10%. In separate experiments, ATP-sensitive potassium channels (KATP) were blocked by extraluminal application of glibenclamide (Glib), Ca2+-activated potassium channels (KCa) by tetraethylammonium (TEA), voltage-gated potassium channels (Kv) by 4-aminopyridine, and inward rectifier potassium channels (KIR) by BaCl2. Na+-K+-ATP-ase was inhibited by ouabain. Application of TEA slightly constricted the arteries at pH 7.4 and slightly but significantly attenuated the vasodilation to acidosis. Inhibition of the other potassium channels or Na+-K+-ATP-ase had no effect. Combined blockade of KATP and KCa channels further reduced resting diameter, and abolished acidosis induced vasodilation. The authors conclude that mainly KCa channels are active under resting conditions. KATP and KCa channels are responsible for vasodilation to acidosis. Activity of one of these potassium channel families is sufficient for vasodilation to acidosis, and only combined inhibition completely abolishes vasodilation. During NO synthase inhibition, dilation to the KATP channel opener pinacidil or the KCa channel opener NS1619 was attenuated or abolished, respectively. The authors suggest that a basal perivascular NO level is necessary for physiologic KATP and KCa channel function in rat MCA. Future studies have to elucidate whether this NO dependent effect on KATP and KCa channel function is a principle mechanism of NO induced modulation of cerebrovascular reactivity and whether the variability of findings in the literature concerning a modulatory role of NO can be explained by different levels of vascular NO/cGMP concentrations within the cerebrovascular tree.
APA, Harvard, Vancouver, ISO, and other styles
50

Zhang, Xiaomeng, Beilei Wang, Zhenzhen Liu, Yubin Zhou, and Lupei Du. "How to Fluorescently Label the Potassium Channel: A Case in hERG." Current Medicinal Chemistry 27, no. 18 (June 3, 2020): 3046–54. http://dx.doi.org/10.2174/0929867326666181129094455.

Full text
Abstract:
hERG (Human ether-a-go-go-related gene) potassium channel, which plays an essential role in cardiac action potential repolarization, is responsible for inherited and druginduced long QT syndrome. Recently, the Cryo-EM structure capturing the open conformation of hERG channel was determined, thus pushing the study on hERG channel at 3.8 Å resolution. This report focuses primarily on summarizing the design rationale and application of several fluorescent probes that target hERG channels, which enables dynamic and real-time monitoring of potassium pore channel affinity to further advance the understanding of the channels.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography