Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
PLoS One ; 13(8): e0201092, 2018.
Article in English | MEDLINE | ID: mdl-30110354

ABSTRACT

We screened a library of botanical compounds purified from plants of Vietnam for modulators of the activity of a two-pore domain K+ channel, TREK-1, and we identified a hydroxycoumarin-related compound, ostruthin, as an activator of this channel. Ostruthin increased whole-cell TREK-1 channel currents in 293T cells at a low concentration (EC50 = 5.3 µM), and also activity of the TREK-2 channel (EC50 = 3.7 mM). In contrast, ostruthin inhibited other K+ channels, e.g. human ether-à-go-go-related gene (HERG1), inward-rectifier (Kir2.1), voltage-gated (Kv1.4), and two-pore domain (TASK-1) at higher concentrations, without affecting voltage-gated potassium channel (KCNQ1 and 3). We tested the effect of this compound on mouse anxiety- and depression-like behaviors and found anxiolytic activity in the open-field, elevated plus maze, and light/dark box tests. Of note, ostruthin also showed antidepressive effects in the forced swim and tail suspension tests, although previous studies reported that inhibition of TREK-1 channels resulted in an antidepressive effect. The anxiolytic and antidepressive effect was diminished by co-administration of a TREK-1 blocker, amlodipine, indicating the involvement of TREK-1 channels. Administration of ostruthin suppressed the stress-induced increase in anti-c-Fos immunoreactivity in the lateral septum, without affecting immunoreactivity in other mood disorder-related nuclei, e.g. the amygdala, paraventricular nuclei, and dorsal raphe nucleus. Ostruthin may exert its anxiolytic and antidepressive effects through a different mechanism from current drugs.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Potassium Channels, Tandem Pore Domain/agonists , Umbelliferones/pharmacology , Amlodipine/pharmacology , Animals , Anxiety/drug therapy , Anxiety/metabolism , Brain/drug effects , Brain/metabolism , Depression/drug therapy , Depression/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/metabolism , Male , Mice, Inbred ICR , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/pharmacology , Phytochemicals/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolism
2.
Pak J Pharm Sci ; 29(5): 1513-1517, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27731805

ABSTRACT

This research is to explore the effects of traditional Chinese medicine Ginseng-spikenard heart-nourishing capsule on the inactivation of c-type Kv1.4 channels (Kv1.4∆N) in Xenopus laevis oocytes with two-electrode voltageclamp technique. Defolliculated oocytes (stage V-VI) were injected with transcribed cRNAs of ferret Kv1.4δN channels. During recording, oocytes were continuously perfused with ND96 solution (control group) and solution prepared from Ginseng-spikenard heart-nourishing capsule (experimental group). Results found that, at the command potential of +50 mV, the current of experimental group was reduced to 48.33±4.0% of that in control group. The inactivation time constants in control and experimental groups were 2962.56±175.35 ms and 304.13±36.22ms, respectively (P<0.05, n=7). The recovery time of fKv1.4∆N channel after inactivation in control group and experimental groups was 987±68.39 ms and 1734.15±98.45 ms, respectively (P<0.05, n=5). Ginseng-spikenard heart-nourishing capsule can inhibit the Kv1.4δN channel, which may be one of the mechanisms of underlying antiarrhythmia.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Kv1.4 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Animals , Female , Ferrets , Gene Transfer Techniques , Kinetics , Kv1.4 Potassium Channel/genetics , Kv1.4 Potassium Channel/metabolism , Membrane Potentials , Oocytes , Xenopus laevis
3.
J Neuroimmunol ; 258(1-2): 61-6, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23561592

ABSTRACT

There are no immunological markers to predict the prognosis of thymoma-associated myasthenia gravis (MG). Clinical and immunological factors associated with thymoma recurrence or MG relapse were examined by logistic analyses in 56 Japanese patients with thymoma-associated MG. Patients with anti-Kv1.4 antibodies showed higher frequencies of thymoma recurrence and MG relapse compared to those without. Anti-Kv1.4 antibody, Masaoka stage 4, World Health Organization type B3, and adjuvant radiotherapy were associated with thymoma recurrence. Multivariate analyses showed that anti-Kv1.4 antibody was the only independent factor associated with MG relapse. Anti-Kv1.4 antibody is a useful predictor of the prognosis of thymoma-associated MG.


Subject(s)
Autoantigens/immunology , Kv1.4 Potassium Channel/immunology , Myasthenia Gravis/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Adolescent , Adult , Asian People , Autoantibodies/blood , Autoantibodies/immunology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Myasthenia Gravis/complications , Prognosis , Recurrence , Thymoma/complications , Thymus Neoplasms/complications , Young Adult
4.
Pharmazie ; 63(6): 475-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18604993

ABSTRACT

As the major component of I(to) (slow), Kv1.4 channel plays an important role in repolarization of cardiac myocytes. C-type inactivation is one of Kv1.4 inactivation and can be affected by open channel blockers. We used the two-electrode voltage clamp technique to observe the effect of amiodarone on Kv1.4 C-type inactivation and compare amiodarone's effects on Kv1.4 with propafenone and verapamil. Our data show that those three antiarrhythmic drugs blocked fKv1.4 delta N (N-terminal deleted Kv1.4 channel from ferret heart) in voltage- and frequent-dependent manners. The amiodarone's IC50 was 489.23 +/- 4.72 microM, higher than that of propafenone (98.97 +/- 1.13 microM) and verapamil (263.26 +/- 6.89 microM) for fKv1.4 delta N channel (+50 mV). After application of amiodarone, propafenone and verapamil, fKv1.4 delta N inactivation becomes bi-exponential: the faster portion of inactivation (drug-induced inactivation) and the slower portion of inactivation (C-type inactivation). Amiodarone and verapamil fastened C-type inactivation in fKv1.4 delta N, but propafenone did not. Unlike propafenone that had no effect on fKv1.4 delta N recovery, amiodarone and verapamil slowed recovery in fKv1.4 delta N.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Kv1.4 Potassium Channel/drug effects , Propafenone/pharmacology , Verapamil/pharmacology , Xenopus Proteins/drug effects , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Electrophysiology , Female , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Xenopus laevis
5.
Eur J Pain ; 12(2): 189-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17584507

ABSTRACT

Voltage-gated K+ (Kv) channels are one of the important physiological regulators of the membrane potentials in excitable cells, including sensory ganglion neurons. The aim of the present study was to investigate whether temporomandibular joint (TMJ) inflammation alters expression of Kv channel subtype 1.4 (Kv1.4) of trigeminal ganglion (TRG) neurons innervating TMJ relating allodynia (pain caused by normally innoxious stimulation), by using both behavioral and immunohistochemical techniques. TMJ inflammation was induced by injection of Complete Freund's Adjuvant (CFA) into the rat TMJ. The threshold for escape from mechanical stimulation applied to the orofacial area in TMJ inflamed rats was significantly lower than that in naïve rats. TMJ afferents were identified by fluorogold (FG) labeling. The mean numbers of Kv1.4-/neurofilament (NF) 200(myelinated fiber marker) positive- and negative-immunoreactivities FG-labeled small-/medium-diameter TRG neurons in inflamed rats were significantly decreased when compared with those in the naïve rats. These findings suggest that TMJ inflammation reduces the expression of Kv1.4 subunits in the small-/medium sized (Adelta-/C-) TRG neurons and this may contribute to trigeminal inflammatory allodynia in TMJ disorder. These results lead us to suggest that Kv channel openers may be a potential therapeutic agents for prevention of mechanical allodynia.


Subject(s)
Arthritis/metabolism , Kv1.4 Potassium Channel/metabolism , Neurons/metabolism , Temporomandibular Joint , Trigeminal Ganglion/metabolism , Animals , Arthritis/chemically induced , Arthritis/pathology , Cell Size , Freund's Adjuvant , Hyperalgesia/chemically induced , Immunohistochemistry , Male , Rats , Rats, Wistar , Temporomandibular Joint/innervation , Trigeminal Ganglion/pathology
6.
J Physiol ; 571(Pt 2): 371-89, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16373387

ABSTRACT

Potassium channels are extremely diverse regulators of neuronal excitability. As part of an investigation into how this molecular diversity is utilized by neurones, we examined the expression and biophysical properties of native Kv1 channels in layer II/III pyramidal neurones from somatosensory and motor cortex. Single-cell RT-PCR, immunocytochemistry, and whole cell recordings with specific peptide toxins revealed that individual pyramidal cells express multiple Kv1 alpha-subunits. The most abundant subunit mRNAs were Kv1.1 > 1.2 > 1.4 > 1.3. All of these subunits were localized to somatodendritic as well as axonal cell compartments. These data suggest variability in the subunit complexion of Kv1 channels in these cells. The alpha-dendrotoxin (alpha-DTX)-sensitive current activated more rapidly and at more negative potentials than the alpha-DTX-insensitive current, was first observed at voltages near action potential threshold, and was relatively insensitive to holding potential. The alpha-DTX-sensitive current comprised about 10% of outward current at steady-state, in response to steps from -70 mV. From -50 mV, this percentage increased to approximately 20%. All cells expressed an alpha-DTX-sensitive current with slow inactivation kinetics. In some cells a transient component was also present. Deactivation kinetics were voltage dependent, such that deactivation was slow at potentials traversed by interspike intervals during repetitive firing. Because of its kinetics and voltage dependence, the alpha-DTX-sensitive current should be most important at physiological resting potentials and in response to brief stimuli. Kv1 channels should also be important at voltages near threshold and corresponding to interspike intervals.


Subject(s)
Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/metabolism , Kv1.4 Potassium Channel/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Animals , Delayed Rectifier Potassium Channels , Elapid Venoms/pharmacology , Immunohistochemistry , Ion Channel Gating/drug effects , Motor Cortex/metabolism , Neocortex/cytology , Neocortex/physiology , Potassium Channel Blockers/pharmacology , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/metabolism
7.
Am J Physiol Heart Circ Physiol ; 288(5): H2077-87, 2005 May.
Article in English | MEDLINE | ID: mdl-15637125

ABSTRACT

Action potential (AP) prolongation is a hallmark of failing myocardium. Functional downregulation of K currents is a prominent feature of cells isolated from failing ventricles. The detailed changes in K current expression differ depending on the species, the region of the heart, and the mechanism of induction of heart failure. We used complementary approaches to study K current downregulation in pacing tachycardia-induced heart failure in the rabbit. The AP duration (APD) at 90% repolarization was significantly longer in cells isolated from failing hearts compared with controls (539 +/- 162 failing vs. 394 +/- 114 control, P < 0.05). The major K currents in the rabbit heart, inward rectifier potassium current (I(K1)), transient outward (I(to)), and delayed rectifier current (I(K)) were functionally downregulated in cells isolated from failing ventricles. The mRNA levels of Kv4.2, Kv1.4, KChIP2, and Kir2.1 were significantly downregulated, whereas the Kv4.3, Erg, KvLQT1, and minK were unaltered in the failing ventricles compared with the control left ventricles. Significant downregulation in the long splice variant of Kv4.3, but not in the total Kv4.3, Kv4.2, and KChIP2 immunoreactive protein, was observed in cells isolated from the failing ventricle with no change in Kv1.4, KvLQT1, and in Kir2.1 immunoreactive protein levels. Multiple cellular and molecular mechanisms underlie the downregulation of K currents in the failing rabbit ventricle.


Subject(s)
Calcium-Binding Proteins/genetics , Heart Failure/physiopathology , Potassium Channels, Voltage-Gated/genetics , Action Potentials/physiology , Animals , Calcium-Binding Proteins/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Female , Gene Expression/physiology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kv Channel-Interacting Proteins , Kv1.4 Potassium Channel , Male , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Voltage-Gated/metabolism , RNA, Messenger/analysis , Rabbits , Shal Potassium Channels
8.
J Biol Chem ; 278(28): 25558-67, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12730233

ABSTRACT

Kv1.4 and Kv1.1 potassium channels are expressed in brain as mature glycoproteins that are trans-Golgi glycosylated. When expressed in cell lines these homomers had very different trans-Golgi glycosylation efficiencies and cell surface expression levels with Kv1.4 > Kv1.1 for both parameters (Zhu, J., Watanabe, I., Gomez, B., and Thornhill, W. B. (2001) J. Biol. Chem. 276, 39419-39427). This previous study identified determinants in the outer pore region of Kv1.4 and Kv1.1 that positively and negatively, respectively, affected these events when expressed as homomers. Here we investigated which subunit exhibited positive or negative effects on these processes when expressed as heteromers. Kv1.4/Kv1.1 heteromers, by coexpression or expression as tandem-linked heteromers, were expressed on the cell surface at approximately 20-fold lower levels versus Kv1.4 homomers but they were trans-Golgi glycosylated. The lower Kv1.4/Kv1.1 expression level was not rescued by Kvbeta 2.1 subunits. Thus Kv1.1 inhibited high cell surface expression and partially retained the heteromer in the endoplasmic reticulum, whereas Kv1.4 stimulated trans-Golgi glycosylation. The subunit determinants and cellular events responsible for these differences were investigated. In a Kv1.4/Kv1.1 heteromer, the Kv1.1 pore was a major negative determinant, and it inhibited high cell surface expression because it induced high partial endoplasmic reticulum retention and it decreased protein stability. Other Kv1.1 regions also inhibited high surface expression of heteromers. The Kv1.1 C terminus induced partial Golgi retention and contributed to a decreased protein stability, whereas the Kv1.1 N terminus contributed to only a decreased protein stability. Thus a neuron may regulate its cell surface K+ channel protein levels by different Kv1 subfamily homomeric and heteromeric combinations that affect intracellular retention characteristics and protein stability.


Subject(s)
Cell Membrane/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/chemistry , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Biotinylation , CHO Cells , COS Cells , Cricetinae , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dimerization , Endoplasmic Reticulum/metabolism , Genes, Dominant , Glycosylation , Golgi Apparatus/metabolism , Immunoblotting , Kv1.1 Potassium Channel , Kv1.4 Potassium Channel , Microscopy, Fluorescence , Molecular Sequence Data , Patch-Clamp Techniques , Plasmids/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Time Factors , Transfection
9.
Am J Physiol Cell Physiol ; 283(4): C1114-21, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12225975

ABSTRACT

Acidosis inhibits current through the Kv1.4 K(+) channel, perhaps as a result of enhancement of C-type inactivation. The mechanism of action of acidosis on C-type inactivation has been studied. A mutant Kv1.4 channel that lacks N-type inactivation (fKv1.4 Delta2-146) was expressed in Xenopus oocytes, and currents were recorded using two-microelectrode voltage clamp. Acidosis increased fKv1.4 Delta2-146 C-type inactivation. Replacement of a pore histidine with cysteine (H508C) abolished the increase. Application of positively charged thiol-specific methanethiosulfonate to fKv1.4 Delta2-146 H508C increased C-type inactivation, mimicking the effect of acidosis. Replacement of a pore lysine with cysteine (K532C) abolished the acidosis-induced increase of C-type inactivation. A model of the Kv1.4 pore, based on the crystal structure of KcsA, shows that H508 and K532 lie close together. It is suggested that the acidosis-induced increase of C-type inactivation involves the charge on H508 and K532.


Subject(s)
Acidosis/metabolism , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Acidosis/chemically induced , Acids/pharmacology , Alkalies/pharmacology , Amino Acid Substitution , Animals , Ferrets , Hydrogen-Ion Concentration/drug effects , Kv1.4 Potassium Channel , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Microinjections , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Engineering , RNA, Complementary/metabolism , Structure-Activity Relationship , Xenopus laevis
10.
J Biol Chem ; 277(5): 3640-6, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11723117

ABSTRACT

The molecular mechanisms underlying the protein assembly at synaptic junctions are thought to be important for neural functions. PSD-95, one of the major postsynaptic density proteins, is composed of three PDZ domains (PDZ1, PDZ2, and PDZ3), an SH3 domain, and a GK (guanylate kinase ) domain. It binds to the N-methyl-D-aspartate glutamate receptor NR2 subunit or to the Shaker-type K(+) channel, Kv1.4, via the PDZ1 or PDZ2 domain, whereas PDZ3 binds to distinct partners. The intramolecular interaction of these multiple domains has been implicated in efficient protein clustering. We introduced missense and deletion mutations into PDZ1 (PDZ1mDelta) and/or PDZ2 (PDZ2mDelta) of the full-length PSD-95 to disrupt the association of each domain with the target proteins, while preserving the overall structure. The ion channel clustering activities of the PSD-95 mutants were analyzed in COS-1 cells coexpressing each mutant and Kv1.4. The mutant bearing the dysfunctional PDZ2 (PSD-95:1-2mDelta) showed significantly reduced clustering efficiency, whereas the mutant with the dysfunctional PDZ1 (PSD-95:1mDelta-2) exhibited activity comparable with the wild-type activity. Furthermore, we also examined the requirements for the position of PDZ2 in full-length PSD-95 by constructing a series of PDZ1-PDZ2 inversion mutants. Surprisingly, the clustering activity of PSD-95:2-1mDelta was severely defective. Taken together, these findings show that PDZ2, which is endowed with the highest affinity for Kv1.4, is required for efficient ligand binding. In addition, the ligand binding at the position of the second PDZ domain in full-length PSD-95 is prerequisite for efficient and typical cluster formation. This study suggests that the correct placement of the multiple domains in the full-length PSD-95 protein is necessary for the optimal protein activity.


Subject(s)
Nerve Tissue Proteins/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , COS Cells , Chlorocebus aethiops , DNA, Complementary , Fluorescent Antibody Technique, Indirect , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Kv1.4 Potassium Channel , Ligands , Mammals , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptide Fragments/chemistry , Polymerase Chain Reaction , Potassium Channels/chemistry , Potassium Channels/genetics , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Transfection , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL