Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
SLAS Discov ; 26(3): 439-449, 2021 03.
Article in English | MEDLINE | ID: mdl-32830616

ABSTRACT

Inhibition of the KCa3.1 potassium channel has therapeutic potential in a variety of human diseases, including inflammation-associated disorders and cancers. However, KCa3.1 inhibitors with high therapeutic promise are currently not available. This study aimed to establish a screening assay for identifying inhibitors of KCa3.1 in native cells and from library compounds derived from natural products in Thailand. The screening platform was successfully developed based on a thallium flux assay in intestinal epithelial (T84) cells with a Z' factor of 0.52. The screening of 1352 compounds and functional validation using electrophysiological analyses identified 8 compounds as novel KCa3.1 inhibitors with IC50 values ranging from 0.14 to 6.57 µM. These results indicate that the assay developed is of excellent quality for high-throughput screening and capable of identifying KCa3.1 inhibitors. This assay may be useful in identifying novel KCa3.1 inhibitors that may have therapeutic potential for inflammation-associated disorders and cancers.


Subject(s)
Biological Products/pharmacology , Epithelial Cells/drug effects , High-Throughput Screening Assays , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Small Molecule Libraries/pharmacology , Thallium/metabolism , Apamin/pharmacology , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , HCT116 Cells , HT29 Cells , Humans , Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Ion Channel Gating/drug effects , Ion Transport , Ouabain/pharmacology , Oximes/pharmacology , Potassium/metabolism , Pyrazoles/pharmacology
2.
J Gastrointestin Liver Dis ; 29(4): 539-547, 2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33331347

ABSTRACT

BACKGROUND AND AIMS: The K + channel KCNN4 is involved in many inflammatory diseases. Previous work has shown that this channel is involved in epithelial ion transport and intestinal restitution. In inflammatory bowel diseases (IBD) a defective epithelial barrier can lead to typical symptoms like secretory diarrhea and the formation of intestinal ulcers. We compared surgical samples from patients with IBD, diverticulitis and controls without inflammation to determine the potential role of KCNN4 as a diagnostic marker and/or therapeutic target. METHODS: mRNA-levels of KCNN4 and a control K + channel were determined in intestinal epithelial cells (IEC) from patients with IBD, diverticulitis and controls. In addition, we performed a Western blot analysis of KCNN4 and a respective control K + channel in IEC from patients with IBD. Furthermore, we determined epithelial barrier integrity by measuring the flux of fluorescent-labeled dextran beads across a cell monolayer upon incubation with interferon-γ. RESULTS: KCNN4 mRNA and protein levels were elevated in IEC from patients with Crohn`s disease (CD) and ulcerative colitis (UC). Of note, KCNN4 was not elevated in non-IBD intestinal inflammatory conditions e.g. diverticulitis. Of clinical relevance, pharmacological KCNN4 channel openers stabilized epithelial barrier function in vitro. Thus, KCNN4 may have a protective role in IBD and constitute a therapeutic target. CONCLUSIONS: Our data demonstrate elevated KCNN4 both at mRNA and protein level in IEC specifically from patients with IBD. Therefore, we conclude that KCNN4 could be used as a novel marker for IBD, especially for the establishment of initial diagnosis. Of therapeutic consequence, we show that pharmacological KCNN4 openers stabilize the epithelial barrier. Thus, KCNN4 might be a novel target to diagnose and treat IBD.


Subject(s)
Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Intestinal Mucosa/metabolism , Adult , Aged , Animals , Benzimidazoles/pharmacology , Benzothiazoles/pharmacology , Case-Control Studies , Cell Line , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Crohn Disease/drug therapy , Crohn Disease/genetics , Female , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intestinal Mucosa/drug effects , Male , Membrane Transport Modulators/pharmacology , Middle Aged , Molecular Targeted Therapy , Permeability , Rats , Up-Regulation
3.
Int J Mol Sci ; 20(17)2019 Aug 23.
Article in English | MEDLINE | ID: mdl-31450834

ABSTRACT

The aim of this study was to investigate the hemodynamic effects of SKA-31, an activator of the small (KCa2.x) and intermediate (KCa3.1) conductance calcium-activated potassium channels, and to evaluate its influence on endothelium-derived hyperpolarization (EDH)-KCa2.3/KCa3.1 type relaxation in isolated endothelium-intact small mesenteric arteries (sMAs) from spontaneously hypertensive rats (SHRs). Functional in vivo and in vitro experiments were performed on SHRs or their normotensive controls, Wistar-Kyoto rats (WKY). SKA-31 (1, 3 and 10 mg/kg) caused a brief decrease in blood pressure and bradycardia in both SHR and WKY rats. In phenylephrine-pre-constricted sMAs of SHRs, SKA-31 (0.01-10 µM)-mediated relaxation was reduced and SKA-31 potentiated acetylcholine-evoked endothelium-dependent relaxation. Endothelium denudation and inhibition of nitric oxide synthase (eNOS) and cyclooxygenase (COX) by the respective inhibitors l-NAME or indomethacin, attenuated SKA-31-mediated vasorelaxation. The inhibition of KCa3.1, KCa2.3, KIR and Na+/K+-ATPase by TRAM-34, UCL1684, Ba2+ and ouabain, respectively, reduced the potency and efficacy of the EDH-response evoked by SKA-31. The mRNA expression of eNOS, prostacyclin synthase, KCa2.3, KCa3.1 and KIR were decreased, while Na+/K+-ATPase expression was increased. Collectively, SKA-31 promoted hypotension and vasodilatation, potentiated agonist-stimulated vasodilation, and maintained KCa2.3/KCa3.1-EDH-response in sMAs of SHR with downstream signaling that involved KIR and Na+/K+-ATPase channels. In view of the importance of the dysfunction of endothelium-mediated vasodilatation in the mechanism of hypertension, application of activators of KCa2.3/KCa3.1 channels such as SKA-31 seem to be a promising avenue in pharmacotherapy of hypertension.


Subject(s)
Benzothiazoles/pharmacology , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Essential Hypertension/genetics , Essential Hypertension/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/agonists , Animals , Blood Pressure/drug effects , Cardiovascular System/physiopathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Essential Hypertension/physiopathology , Rats , Rats, Inbred SHR
4.
Int J Mol Sci ; 20(14)2019 Jul 16.
Article in English | MEDLINE | ID: mdl-31315169

ABSTRACT

Systemic hypertension is a major risk factor for the development of cardiovascular disease and is often associated with endothelial dysfunction. KCa2.3 and KCa3.1 channels are expressed in the vascular endothelium and contribute to stimulus-evoked vasodilation. We hypothesized that acute treatment with SKA-31, a selective activator of KCa2.x and KCa3.1 channels, would improve endothelium-dependent vasodilation and transiently lower mean arterial pressure (MAP) in male, spontaneously hypertensive rats (SHRs). Isolated vascular preparations exhibited impaired vasodilation in response to bradykinin (i.e., endothelial dysfunction) compared with Wistar controls, which was associated with decreased bradykinin receptor expression in mesenteric arteries. In contrast, similar levels of endothelial KCa channel expression were observed, and SKA-31 evoked vasodilation was comparable in vascular preparations from both strains. Addition of a low concentration of SKA-31 (i.e., 0.2-0.3 µM) failed to augment bradykinin-induced vasodilation in arteries from SHRs. However, responses to acetylcholine were enhanced. Surprisingly, acute bolus administration of SKA-31 in vivo (30 mg/kg, i.p. injection) modestly elevated MAP compared with vehicle injection. In summary, pharmacological targeting of endothelial KCa channels in SHRs did not readily reverse endothelial dysfunction in situ, or lower MAP in vivo. SHRs thus appear to be less responsive to endothelial KCa channel activators, which may be related to their vascular pathology.


Subject(s)
Endothelium, Vascular/drug effects , Hypertension/drug therapy , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Acetylcholine/pharmacology , Animals , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Blood Pressure , Bradykinin/pharmacology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hypertension/physiopathology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Rats , Rats, Inbred SHR , Rats, Wistar , Receptors, Bradykinin/genetics , Receptors, Bradykinin/metabolism , Vasodilation
5.
Science ; 360(6388): 508-513, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29724949

ABSTRACT

Small-conductance Ca2+-activated K+ (SK) channels mediate neuron excitability and are associated with synaptic transmission and plasticity. They also regulate immune responses and the size of blood cells. Activation of SK channels requires calmodulin (CaM), but how CaM binds and opens SK channels has been unclear. Here we report cryo-electron microscopy (cryo-EM) structures of a human SK4-CaM channel complex in closed and activated states at 3.4- and 3.5-angstrom resolution, respectively. Four CaM molecules bind to one channel tetramer. Each lobe of CaM serves a distinct function: The C-lobe binds to the channel constitutively, whereas the N-lobe interacts with the S4-S5 linker in a Ca2+-dependent manner. The S4-S5 linker, which contains two distinct helices, undergoes conformational changes upon CaM binding to open the channel pore. These structures reveal the gating mechanism of SK channels and provide a basis for understanding SK channel pharmacology.


Subject(s)
Calmodulin/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/chemistry , Acetamides/chemistry , Calmodulin/agonists , Calmodulin/ultrastructure , Cryoelectron Microscopy , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/ultrastructure , Potassium Channel Blockers/chemistry , Protein Domains , Thiazines/chemistry , Trityl Compounds/chemistry
6.
Eur J Pharmacol ; 831: 60-67, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29753043

ABSTRACT

It is now well recognized that endothelial KCa2.3 and KCa3.1 channel activities contribute to dilation of resistance arteries via endothelium-mediated hyperpolarization and vascular smooth muscle relaxation. In this study, we have investigated the functional effect of the KCa channel activator SKA-31 in third order rat mesenteric arteries using arterial pressure myography. Isolated arteries were cannulated, pressurized intraluminally to 70 mmHg at 36 °C and then constricted with 1 µM phenylephrine. Acute bath exposure to SKA-31 evoked a robust and reversible inhibition of developed tone (IC50 = 0.22 µM). The vasodilatory effects of SKA-31 and acetylcholine were blunted in the presence of KCa2.3 and KCa3.1 channel antagonists, and were largely prevented following endothelial denudation. Western blot and q-PCR analyses of isolated mesenteric arteries revealed KCa2.3 and KCa3.1 channel expression at the protein and mRNA levels, respectively. Penitrem-A, an inhibitor of KCa1.1 channels, decreased vasodilatory responses to acetylcholine, sodium nitroprusside and NS-1619, but had little effect on SKA-31. Similarly, bath exposure to the eNOS inhibitor L-NAME did not alter SKA-31 and acetylcholine-mediated vasodilation. Collectively, these data highlight the major cellular mechanisms by which the endothelial KCa channel activator SKA-31 inhibits agonist-evoked vasoconstriction in rat small mesenteric arteries.


Subject(s)
Benzothiazoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Mesenteric Arteries/drug effects , Small-Conductance Calcium-Activated Potassium Channels/agonists , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Dose-Response Relationship, Drug , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mesenteric Arteries/metabolism , Myography , Rats, Sprague-Dawley , Signal Transduction/drug effects , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism
7.
Mol Pharmacol ; 92(4): 469-480, 2017 10.
Article in English | MEDLINE | ID: mdl-28760780

ABSTRACT

Intermediate-conductance (KCa3.1) and small-conductance (KCa2) calcium-activated K+ channels are gated by calcium binding to calmodulin (CaM) molecules associated with the calmodulin-binding domain (CaM-BD) of these channels. The existing KCa activators, such as naphtho[1,2-d]thiazol-2-ylamine (SKA-31), 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), and 1-ethylbenzimidazolin-2-one (EBIO), activate both channel types with similar potencies. In a previous chemistry effort, we optimized the benzothiazole pharmacophore of SKA-31 toward KCa3.1 selectivity and identified 5-methylnaphtho[2,1-d]oxazol-2-amine (SKA-121), which exhibits 40-fold selectivity for KCa3.1 over KCa2.3. To understand why introduction of a single CH3 group in five-position of the benzothiazole/oxazole system could achieve such a gain in selectivity for KCa3.1 over KCa2.3, we first localized the binding site of the benzothiazoles/oxazoles to the CaM-BD/CaM interface and then used computational modeling software to generate models of the KCa3.1 and KCa2.3 CaM-BD/CaM complexes with SKA-121. Based on a combination of mutagenesis and structural modeling, we suggest that all benzothiazole/oxazole-type KCa activators bind relatively "deep" in the CaM-BD/CaM interface and hydrogen bond with E54 on CaM. In KCa3.1, SKA-121 forms an additional hydrogen bond network with R362. In contrast, NS309 sits more "forward" and directly hydrogen bonds with R362 in KCa3.1. Mutating R362 to serine, the corresponding residue in KCa2.3 reduces the potency of SKA-121 by 7-fold, suggesting that R362 is responsible for the generally greater potency of KCa activators on KCa3.1. The increase in SKA-121's KCa3.1 selectivity compared with its parent, SKA-31, seems to be due to better overall shape complementarity and hydrophobic interactions with S372 and M368 on KCa3.1 and M72 on CaM at the KCa3.1-CaM-BD/CaM interface.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Ion Channel Gating/physiology , Oxazoles/metabolism , Oxazoles/pharmacology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Ion Channel Gating/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
8.
Sci Rep ; 7(1): 4340, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28659615

ABSTRACT

Disorders in cell signaling mediated by ATP or histamine, activating specific membrane receptors, have been frequently associated with tumorigenesis. Among the elements of response to purinergic (and histaminergic) signaling, ion channel activation controls essential cellular processes in cancer, such as cell proliferation, motility, and death. Here, we studied the effects that ATP had on electrical properties of human ovarian adenocarcinoma cells named SKOV-3. ATP caused increase in intracellular Ca2+ concentration ([Ca2+]i) and, concurrently, it evoked a complex electrical response with a conspicuous outward component. This current was generated through P2Y2 receptor activation and opening of K+ channels, KCa3.1, as indicated by electrophysiological and pharmacological analysis, as well as by immunodetection and specific silencing of P2Y2 or KCa3.1 gene by esiRNA transfection. Low µM ATP concentration increased SKOV-3 cell migration, which was strongly inhibited by KCa3.1 channel blockers and by esiRNA-generated P2Y2 or KCa3.1 downregulation. Finally, in human ovarian tumors, the P2Y2 and KCa3.1 proteins are expressed and co-localized in neoplastic cells. Thus, stimulation of P2Y2 receptors expressed in SKOV-3 cells promotes motility through KCa3.1 activation. Since P2Y2 and KCa3.1 are co-expressed in primary tumors, our findings suggest that they may play a role in cancer progression.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Ion Channel Gating , Receptors, Purinergic P2Y2/metabolism , Adenosine Triphosphate/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Dose-Response Relationship, Drug , Female , Gene Expression , Gene Silencing , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ions/metabolism , Membrane Potentials , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Potassium Channel Blockers/pharmacology , RNA, Small Interfering/genetics
9.
Vascul Pharmacol ; 79: 24-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26239885

ABSTRACT

SKA-31, an activator of endothelial KCa2.3 and KCa3.1 channels, reduces systemic blood pressure in mice and dogs, however, its effects in larger mammals are not well known. We therefore examined the hemodynamic effects of SKA-31, along with sodium nitroprusside (SNP), in anesthetized, juvenile male domestic pigs. Experimentally, continuous measurements of left ventricular (LV), aortic and inferior vena cava (IVC) pressures, along with flows in the ascending aorta, carotid artery, left anterior descending coronary artery and renal artery, were performed during acute administration of SKA-31 (0.1, 0.3, 1.0, 3.0 and 5.0mg/ml/kg) and a single dose of SNP (5.0 µg/ml/kg). SKA-31 dose-dependently reduced mean aortic pressure (mPAO), with the highest dose decreasing mPAO to a similar extent as SNP (-23 ± 3 and -28 ± 4 mmHg, respectively). IVC pressure did not change. Systemic conductance and conductance in coronary and carotid arteries increased in response to SKA-31 and SNP, but renal artery conductance was unaffected. There was no change in either LV stroke volume (SV) or heart rate (versus the preceding control) for any infusion. With no change in SV, drug-evoked decreases in LV stroke work (SW) were attributed to reductions in mPAO (SW vs. mPAO, r(2)=0.82, P<0.001). In summary, SKA-31 dose-dependently reduced mPAO by increasing systemic and arterial conductances. Primary reductions in mPAO by SKA-31 largely account for associated decreases in SW, implying that SKA-31 does not directly impair cardiac contractility.


Subject(s)
Arterial Pressure/drug effects , Benzothiazoles/pharmacology , Endothelium, Vascular/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/agonists , Animals , Arterial Pressure/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Sus scrofa , Swine
10.
J Am Heart Assoc ; 4(8): e002062, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26304940

ABSTRACT

BACKGROUND: Diabetes is associated with coronary arteriolar endothelial dysfunction. We investigated the role of the small/intermediate (SK(Ca)/IK(Ca)) conductance of calcium-activated potassium channels in diabetes-related endothelial dysfunction. METHODS AND RESULTS: Coronary arterioles (80 to 150 µm in diameter) were dissected from discarded right atrial tissues of diabetic (glycosylated hemoglobin = 9.6±0.25) and nondiabetic patients (glycosylated hemoglobin 5.4±0.12) during coronary artery bypass graft surgery (n=8/group). In-vitro relaxation response of precontracted arterioles was examined in the presence of the selective SK(Ca)/IK(Ca) activator NS309 and other vasodilatory agents. The channel density and membrane potential of diabetic and nondiabetic endothelial cells was measured by using the whole cell patch-clamp technique. The protein expression and distribution of the SK(Ca)/IK(Ca) in the human myocardium and coronary arterioles was examined by Western blotting and immunohistochemistry. Our results indicate that diabetes significantly reduced the coronary arteriolar response to the SK(Ca)/IK(Ca) activator NS309 compared to the respective responses of nondiabetic vessels (P<0.05 versus nondiabetes). The relaxation response of diabetic arterioles to NS309 was prevented by denudation of endothelium (P=0.001 versus endothelium-intact). Diabetes significantly decreased endothelial SK(Ca)/IK(Ca) currents and hyperpolarization induced by the SK(Ca)/IK(Ca) activator NS309 as compared with that of nondiabetics. There were no significant differences in the expression and distribution of SK(Ca)/IK(Ca) proteins in the coronary microvessels. CONCLUSIONS: Diabetes is associated with inactivation of endothelial SK(Ca)/IK(Ca) channels, which may contribute to endothelial dysfunction in diabetic patients.


Subject(s)
Arterioles/metabolism , Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Diabetic Angiopathies/metabolism , Endothelial Cells/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Vasodilation , Aged , Aged, 80 and over , Arterioles/drug effects , Arterioles/physiopathology , Biomarkers/blood , Case-Control Studies , Cells, Cultured , Coronary Artery Disease/diagnosis , Coronary Artery Disease/physiopathology , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/physiopathology , Dose-Response Relationship, Drug , Down-Regulation , Endothelial Cells/drug effects , Female , Glycated Hemoglobin/analysis , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Male , Membrane Potentials , Middle Aged , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/agonists , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
J Cardiovasc Pharmacol ; 66(1): 118-27, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25815673

ABSTRACT

Endothelial KCa2.3 and KCa3.1 channels contribute to the regulation of myogenic tone in resistance arteries by Ca(2+)-mobilizing vasodilatory hormones. To define further the functional role of these channels in distinct vascular beds, we have examined the vasodilatory actions of the KCa channel activator SKA-31 in myogenically active rat cremaster and middle cerebral arteries. Vessels pressurized to 70 mm Hg constricted by 80-100 µm (ie, 25%-45% of maximal diameter). SKA-31 (10 µM) inhibited myogenic tone by 80% in cremaster and ∼65% in middle cerebral arteries, with IC50 values of ∼2 µM in both vessels. These vasodilatory effects were largely prevented by the KCa2.3 blocker UCL1684 and the KCa3.1 blocker TRAM-34 and abolished by endothelial denudation. Preincubation with N(G) nitro L-arginine methyl ester (L-NAME, 0.1 mM) did not affect the inhibitory response to SKA-31, but attenuated the ACh-evoked dilation by ∼45%. Penitrem-A, a blocker of BK(Ca) channels, did not alter SKA-31 evoked vasodilation but did reduce the inhibition of myogenic tone by ACh, the BKCa channel activator NS1619, and sodium nitroprusside. Collectively, these data demonstrate that SKA-31 produces robust inhibition of myogenic tone in resistance arteries isolated from distinct vascular beds in an endothelium-dependent manner.


Subject(s)
Benzothiazoles/pharmacology , Cerebral Arteries/drug effects , Endothelium, Vascular/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/agonists , Vasodilation/drug effects , Animals , Cerebral Arteries/physiology , Endothelium, Vascular/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Male , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/physiology , Vasodilation/physiology
12.
Transl Res ; 166(1): 89-102, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25583444

ABSTRACT

The role of ion channels is largely unknown in chemokine-induced migration in nonexcitable cells such as dendritic cells (DCs). Here, we examined the role of intermediate-conductance calcium-activated potassium channel (KCa3.1) and chloride channel (CLC3) in lymphatic chemokine-induced migration of DCs. The amplitude and kinetics of chemokine ligand (CCL19/CCL21)-induced Ca(2+) influx were associated with chemokine receptor 7 expression levels, extracellular-free Ca(2+) and Cl(-), and independent of extracellular K(+). Chemokines (CCL19 and CCL21) and KCa3.1 activator (1-ethyl-1,3-dihydro-2H-benzimidazol-2-one) induced plasma membrane hyperpolarization and K(+) efflux, which was blocked by 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole, suggesting that KCa3.1 carried larger conductance than the inward calcium release-activated calcium channel. Blockade of KCa3.1, low Cl(-) in the medium, and low dose of 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS) impaired CCL19/CCL21-induced Ca(2+) influx, cell volume change, and DC migration. High doses of DIDS completely blocked DC migration possibly by significantly disrupting mitochondrial membrane potential. In conclusion, KCa3.1 and CLC3 are critical in human DC migration by synergistically regulating membrane potential, chemokine-induced Ca(2+) influx, and cell volume.


Subject(s)
Chloride Channels/metabolism , Dendritic Cells/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Benzimidazoles/pharmacology , Calcium Signaling , Cell Movement/drug effects , Cell Movement/immunology , Cell Movement/physiology , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Chloride Channels/antagonists & inhibitors , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Membrane Potentials/drug effects , Pyrazoles/pharmacology , Receptors, CCR7/metabolism , Translational Research, Biomedical
13.
PLoS One ; 9(10): e109432, 2014.
Article in English | MEDLINE | ID: mdl-25302606

ABSTRACT

BACKGROUND AND AIMS: Endothelial small- and intermediate-conductance KCa channels, SK3 and IK1, are key mediators in the endothelium-derived hyperpolarization and relaxation of vascular smooth muscle and also in the modulation of endothelial Ca2+ signaling and nitric oxide (NO) release. Obesity is associated with endothelial dysfunction and impaired relaxation, although how obesity influences endothelial SK3/IK1 function is unclear. Therefore we assessed whether the role of these channels in the coronary circulation is altered in obese animals. METHODS AND RESULTS: In coronary arteries mounted in microvascular myographs, selective blockade of SK3/IK1 channels unmasked an increased contribution of these channels to the ACh- and to the exogenous NO- induced relaxations in arteries of Obese Zucker Rats (OZR) compared to Lean Zucker Rats (LZR). Relaxant responses induced by the SK3/IK1 channel activator NS309 were enhanced in OZR and NO- endothelium-dependent in LZR, whereas an additional endothelium-independent relaxant component was found in OZR. Fura2-AM fluorescence revealed a larger ACh-induced intracellular Ca2+ mobilization in the endothelium of coronary arteries from OZR, which was inhibited by blockade of SK3/IK1 channels in both LZR and OZR. Western blot analysis showed an increased expression of SK3/IK1 channels in coronary arteries of OZR and immunohistochemistry suggested that it takes place predominantly in the endothelial layer. CONCLUSIONS: Obesity may induce activation of adaptive vascular mechanisms to preserve the dilator function in coronary arteries. Increased function and expression of SK3/IK1 channels by influencing endothelial Ca2+ dynamics might contribute to the unaltered endothelium-dependent coronary relaxation in the early stages of obesity.


Subject(s)
Calcium Signaling/physiology , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Obesity/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Up-Regulation , Animals , Calcium Signaling/drug effects , Coronary Vessels/drug effects , Endothelium, Vascular/drug effects , Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Myography , Obesity/genetics , Oximes/pharmacology , Rats , Rats, Zucker , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/genetics , Vasodilation/drug effects , Vasodilation/physiology
14.
Chin J Physiol ; 57(3): 137-51, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24826782

ABSTRACT

Nitric oxide (NO) is produced by nitric oxide synthase (NOS) in dermal fibroblasts and is important during wound healing. Intermediate conductance Ca²âº-activated K+ (IK; IK1; KCa3.1; IKCa; SK4; KCNN4) channels contribute to NOS upregulation, NO production, and various NO-mediated essential functions in many kinds of cells. To determine if the action of NO is linked to IK channel regulation in human dermal fibroblasts, we investigated the expression of IK channels in the cells and the effects and mechanisms of NO on the channels using RT-PCR, western blot analysis, immunocytochemistry and whole-cell and single-channel patch-clamp techniques. The presence of functional IK channels at the RNA, protein and membrane levels was demonstrated and S-nitroso-N-acetylpenicillamine (SNAP) was shown to significantly increase IK currents. The effects of NO were abolished by pretreatment with KT5823 or 1H-[1,2,4]-oxadiazolo [4,3-a]quinoxalin-1-one (ODQ) but not with KT5720. In addition, IK currents were increased by protein kinase G1α or 8-bromo-cGMP but not by forskolin, 8-bromo-cAMP, or catalytic subunits of protein kinase A (PKAcs). On the other hand, PKAcs with cGMP did not increase IK currents, and pretreatment with KT5720 did not block the stimulating effects of 8-Br-cGMP on the IK channels. These data suggest that NO activates IK channels through the PKG but not the PKA pathways, and it seems there is no cross activation between PKG and PKA pathways in human dermal fibroblasts.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Nitric Oxide/pharmacology , Wound Healing/physiology , 4-Aminopyridine/pharmacology , Calcium/metabolism , Cells, Cultured , Colforsin/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dermis/cytology , Dermis/physiology , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Quinoxalines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Toxins, Biological/pharmacology
15.
Arterioscler Thromb Vasc Biol ; 34(1): 127-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24177326

ABSTRACT

OBJECTIVE: Intermediate and small conductance KCa channels IK1 (KCa3.1) and SK3 (KCa2.3) are primary targets of endothelial Ca(2+) signals in the arterial vasculature, and their ablation results in increased arterial tone and hypertension. Activation of IK1 channels by local Ca(2+) transients from internal stores or plasma membrane channels promotes arterial hyperpolarization and vasodilation. Here, we assess arteries from genetically altered IK1 knockout mice (IK1(-/-)) to determine whether IK1 channels exert a positive feedback influence on endothelial Ca(2+) dynamics. APPROACH AND RESULTS: Using confocal imaging and custom data analysis software, we found that although the occurrence of basal endothelial Ca(2+) dynamics was not different between IK1(-/-) and wild-type mice (P>0.05), the frequency of acetylcholine-stimulated (2 µmol/L) Ca(2+) dynamics was greatly decreased in IK1(-/-) endothelium (515±153 versus 1860±319 events; P<0.01). In IK1(-/-)/SK3(T/T) mice, ancillary suppression (+Dox) or overexpression (-Dox) of SK3 channels had little additional effect on the occurrence of events under basal or acetylcholine-stimulated conditions. However, SK3 overexpression did restore the decreased event amplitudes. Removal of extracellular Ca(2+) reduced acetylcholine-induced Ca(2+) dynamics to the same level in wild-type and IK1(-/-) arteries. Blockade of IK1 and SK3 with the combination of charybdotoxin (0.1 µmol/L) and apamin (0.5 µmol/L) or transient receptor potential vanilloid 4 channels with HC-067047 (1 µmol/L) reduced acetylcholine Ca(2+) dynamics in wild-type arteries to the level of IK1(-/-)/SK3(T/T)+Dox arteries. These drug effects were not additive. CONCLUSIONS: IK1, and to some extent SK3, channels exert a substantial positive feedback influence on endothelial Ca(2+) dynamics.


Subject(s)
Acetylcholine/pharmacology , Calcium Signaling/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Feedback, Physiological , Female , Image Processing, Computer-Assisted , Intermediate-Conductance Calcium-Activated Potassium Channels/deficiency , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Kinetics , Male , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/deficiency , Small-Conductance Calcium-Activated Potassium Channels/genetics , Software , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
16.
Assay Drug Dev Technol ; 11(9-10): 551-60, 2013.
Article in English | MEDLINE | ID: mdl-24351043

ABSTRACT

The intermediate-conductance Ca(2+)-activated K(+) channel KCa3.1 (also known as KCNN4, IK1, or the Gárdos channel) plays an important role in the activation of T and B cells, mast cells, macrophages, and microglia by regulating membrane potential, cellular volume, and calcium signaling. KCa3.1 is further involved in the proliferation of dedifferentiated vascular smooth muscle cells and fibroblast and endothelium-derived hyperpolarization responses in the vascular endothelium. Accordingly, KCa3.1 inhibitors are therapeutically interesting as immunosuppressants and for the treatment of a wide range of fibroproliferative disorders, whereas KCa3.1 activators constitute a potential new class of endothelial function preserving antihypertensives. Here, we report the development of QPatch assays for both KCa3.1 inhibitors and activators. During assay optimization, the Ca(2+) sensitivity of KCa3.1 was studied using varying intracellular Ca(2+) concentrations. A free Ca(2+) concentration of 1 µM was chosen to optimally test inhibitors. To identify activators, which generally act as positive gating modulators, a lower Ca(2+) concentration (∼200 nM) was used. The QPatch results were benchmarked against manual patch-clamp electrophysiology by determining the potency of several commonly used KCa3.1 inhibitors (TRAM-34, NS6180, ChTX) and activators (EBIO, riluzole, SKA-31). Collectively, our results demonstrate that the QPatch provides a comparable but much faster approach to study compound interactions with KCa3.1 channels in a robust and reliable assay.


Subject(s)
Biological Assay/methods , Flow Cytometry/methods , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Membrane Potentials/physiology , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , Drug Evaluation, Preclinical/methods , Flow Injection Analysis/methods , HEK293 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Robotics/methods
17.
Biochem Pharmacol ; 85(10): 1486-94, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23500546

ABSTRACT

The proliferation of cardiac fibroblasts is implicated in the pathogenesis of myocardial remodeling and fibrosis. Intermediate-conductance calcium-activated K⁺ channels (K(Ca)3.1 channels) have important roles in cell proliferation. However, it is unknown whether angiotensin II (Ang II), a potent profibrotic molecule, would regulate K(Ca)3.1 channels in cardiac fibroblasts and participate in cell proliferation. In the present study, we investigated whether K(Ca)3.1 channels were regulated by Ang II, and how the channel activity mediated cell proliferation in cultured adult rat cardiac fibroblasts using electrophysiology and biochemical approaches. It was found that mRNA, protein, and current density of K(Ca)3.1 channels were greatly enhanced in cultured cardiac fibroblasts treated with 1 µM Ang II, and the effects were countered by the angiotensin type 1 receptor (AT1R) blocker losartan, the p38-MAPK inhibitor SB203580, the ERK1/2 inhibitor PD98059, and the PI3K/Akt inhibitor LY294002. Ang II stimulated cell proliferation and the effect was antagonized by the K(Ca)3.1 blocker TRAM-34 and siRNA targeting K(Ca)3.1. In addition, Ang II-induced increase of K(Ca)3.1 expression was attenuated by transfection of activator protein-1 (AP-1) decoy oligodeoxynucleotides. These results demonstrate for the first time that Ang II stimulates cell proliferation mediated by upregulating K(Ca)3.1 channels via interacting with the AT1R and activating AP-1 complex through ERK1/2, p38-MAPK and PI3K/Akt signaling pathways in cultured adult rat cardiac fibroblasts.


Subject(s)
Angiotensin II/pharmacology , Fibroblasts/drug effects , Heart Ventricles/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Vasoconstrictor Agents/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cell Proliferation/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Heart Ventricles/cytology , Heart Ventricles/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Losartan/pharmacology , Male , Patch-Clamp Techniques , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Clin Exp Pharmacol Physiol ; 39(7): 608-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22540516

ABSTRACT

1. High dietary Na(+) is associated with impaired vascular endothelial function. However, the underlying mechanisms are not completely understood. In the present study, we investigated whether the endothelial hyperpolarization response to acetylcholine (ACh) exhibited any abnormalities in Wistar rats fed a high-salt diet (HSD) for 1 month and, if so, whether chronic treatment with the angiotensin-converting enzyme inhibitor enalapril or the anti-oxidant tempol could normalize the response. Membrane potential was recorded using the perforated patch-clamp technique on the endothelium of rat aorta. 2. Acetylcholine (2 µmol/L) produced a hyperpolarization sensitive to TRAM-34, a blocker of intermediate-conductance Ca(2+) -sensitive K(+) channels (IK(Ca)), but not to apamin, a blocker of small-conductance Ca(2+)-sensitive K(+) channels (SK(Ca)). NS309 (3 µmol/L), an activator of SK(Ca) and IK(Ca) channels, produced a hyperpolarization of similar magnitude as ACh. 3. In the HSD group, the ACh-evoked hyperpolarization was significantly attenuated compared with that in the control group, which was fed normal chow rather than an HSD. Similarly, the hyperpolarization produced by NS309 was weaker in tissues from HSD-fed rats. 4. Combination of HSD with chronic enalapril treatment (20 mg/kg per day for 1 month) normalized endothelial hyperpolarizing responses to ACh. Chronic tempol treatment (1 mmol/L in tap water for 1 month) prevented the reduced hyperpolarization to ACh. 5. The results of the present study indicate that excess in dietary Na(+) results in a failure of endothelial cells to generate normal IK(Ca) channel-mediated hyperpolarizing responses. Our observations implicate oxidative stress mediated by increased angiotensin II signalling as a mechanism underlying altered endothelial hyperpolarization during dietary salt loading.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antioxidants/pharmacology , Cyclic N-Oxides/pharmacology , Enalapril/pharmacology , Endothelium, Vascular/drug effects , Membrane Potentials/drug effects , Sodium, Dietary/administration & dosage , Acetylcholine/pharmacology , Animals , Apamin/pharmacology , Cholinergic Agonists/pharmacology , Endothelium, Vascular/physiology , Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Oximes/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Spin Labels
19.
Br J Pharmacol ; 167(1): 37-47, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22506557

ABSTRACT

BACKGROUND AND PURPOSE: Small (K(Ca) 2) and intermediate (K(Ca) 3.1) conductance calcium-activated potassium channels (K(Ca) ) may contribute to both epithelium- and endothelium-dependent relaxations, but this has not been established in human pulmonary arteries and bronchioles. Therefore, we investigated the expression of K(Ca) 2.3 and K(Ca) 3.1 channels, and hypothesized that activation of these channels would produce relaxation of human bronchioles and pulmonary arteries. EXPERIMENTAL APPROACH: Channel expression and functional studies were conducted in human isolated small pulmonary arteries and bronchioles. K(Ca) 2 and K(Ca) 3.1 currents were examined in human small airways epithelial (HSAEpi) cells by whole-cell patch clamp techniques. RESULTS: While K(Ca) 2.3 expression was similar, K(Ca) 3.1 protein was more highly expressed in pulmonary arteries than bronchioles. Immunoreactive K(Ca) 2.3 and K(Ca) 3.1 proteins were found in both endothelium and epithelium. K(Ca) currents were present in HSAEpi cells and sensitive to the K(Ca) 2.3 blocker UCL1684 and the K(Ca) 3.1 blocker TRAM-34. In pulmonary arteries contracted by U46619 and in bronchioles contracted by histamine, the K(Ca) 2.3/ K(Ca) 3.1 activator, NS309, induced concentration-dependent relaxations. NS309 was equally potent in relaxing pulmonary arteries, but less potent in bronchioles, than salbutamol. NS309 relaxations were blocked by the K(Ca) 2 channel blocker apamin, while the K(Ca) 3.1 channel blocker, charybdotoxin failed to reduce relaxation to NS309 (0.01-1 µM). CONCLUSIONS AND IMPLICATIONS: K(Ca) 2.3 and K(Ca) 3.1 channels are expressed in the endothelium of human pulmonary arteries and epithelium of bronchioles. K(Ca) 2.3 channels contributed to endo- and epithelium-dependent relaxations suggesting that these channels are potential targets for treatment of pulmonary hypertension and chronic obstructive pulmonary disease.


Subject(s)
Bronchioles/drug effects , Endothelium/drug effects , Epithelial Cells/drug effects , Indoles/pharmacology , Oximes/pharmacology , Pulmonary Artery/drug effects , Small-Conductance Calcium-Activated Potassium Channels/physiology , Aged , Bronchioles/physiology , Cells, Cultured , Endothelium/physiology , Epithelial Cells/physiology , Humans , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Middle Aged , Pulmonary Artery/physiology , RNA, Messenger/metabolism , Respiratory Mucosa/cytology , Small-Conductance Calcium-Activated Potassium Channels/agonists
20.
Am J Respir Cell Mol Biol ; 45(5): 962-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21493782

ABSTRACT

Migration to draining lymph nodes is a critical requirement for dendritic cells (DCs) to control T-cell-mediated immunity. The calcium-activated potassium channel KCa3.1 has been shown to be involved in regulating cell migration in multiple cell types. In this study, KCa3.1 expression and its functional role in lung DC migration were examined. Fluorescence-labeled antigen was intranasally delivered into mouse lungs to label lung Ag-carrying DCs. Lung CD11c(high)CD11b(low) and CD11c(low)CD11b(high) DCs from PBS-treated and ovalbumin (OVA)-sensitized mice were sorted using MACS and FACS. Indo-1 and DiBAC4(3) were used to measure intracellular Ca(2+) and membrane potential, respectively. The mRNA expression of KCa3.1 was examined using real-time PCR. Expression of KCa3.1 protein and CCR7 was measured using flow cytometry. Migration of two lung DC subsets to lymphatic chemokines was examined using TransWell in the absence or presence of the KCa3.1 blocker TRAM-34. OVA sensitization up-regulated mRNA and protein expression of KCa3.1 in lung DCs, with a greater response by the CD11c(high)CD11b(low) than CD11c(low)CD11b(high) DCs. Although KCa3.1 expression in Ag-carrying DCs was higher than that in non-Ag-carrying DCs in OVA-sensitized mice, the difference was not as prominent. However, Ag-carrying lung DCs expressed significantly higher CCR7 than non-Ag-carrying DCs. CCL19, CCL21, and KCa3.1 activator 1-EBIO induced an increase in intracellular calcium in both DC subsets. In addition, 1-EBIO-induced calcium increase was suppressed by TRAM-34. In vitro blockade of KCa3.1 with TRAM-34 impaired CCL19/CCL21-induced transmigration. In conclusion, KCa3.1 expression in lung DCs is up-regulated by OVA sensitization in both lung DC subsets, and KCa3.1 is involved in lung DC migration to lymphatic chemokines.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Lung/immunology , Animals , Benzimidazoles/pharmacology , CD11 Antigens/immunology , Calcium/metabolism , Cell Movement/drug effects , Chemokines/immunology , Dendritic Cells/drug effects , Female , Flow Cytometry , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Lung/drug effects , Membrane Potentials/drug effects , Membrane Potentials/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Pyrazoles/pharmacology , Receptors, CCR7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...