Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L822-L834, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28283479

ABSTRACT

Kv7 potassium channels have recently been found to be expressed and functionally important for relaxation of airway smooth muscle. Previous research suggests that native Kv7 currents are inhibited following treatment of freshly isolated airway smooth muscle cells with bronchoconstrictor agonists, and in intact airways inhibition of Kv7 channels is sufficient to induce bronchiolar constriction. However, the mechanism by which Kv7 currents are inhibited by bronchoconstrictor agonists has yet to be elucidated. In the present study, native Kv7 currents in cultured human trachealis smooth muscle cells (HTSMCs) were observed to be inhibited upon treatment with histamine; inhibition of Kv7 currents was associated with membrane depolarization and an increase in cytosolic Ca2+ ([Ca2+]cyt). The latter response was inhibited by verapamil, a blocker of L-type voltage-sensitive Ca2+ channels (VSCCs). Protein kinase C (PKC) has been implicated as a mediator of bronchoconstrictor actions, although the targets of PKC are not clearly established. We found that histamine treatment significantly and dose-dependently suppressed currents through overexpressed wild-type human Kv7.5 (hKv7.5) channels in cultured HTSMCs, and this effect was inhibited by the PKC inhibitor Ro-31-8220 (3 µM). The PKC-dependent suppression of hKv7.5 currents corresponded with a PKC-dependent increase in hKv7.5 channel phosphorylation. Knocking down or inhibiting PKCα, or mutating hKv7.5 serine 441 to alanine, abolished the inhibitory effects of histamine on hKv7.5 currents. These findings provide the first evidence linking PKC activation to suppression of Kv7 currents, membrane depolarization, and Ca2+ influx via L-type VSCCs as a mechanism for histamine-induced bronchoconstriction.


Subject(s)
Bronchoconstrictor Agents/pharmacology , Histamine/pharmacology , KCNQ Potassium Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Kinase C/metabolism , Trachea/cytology , Calcium/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Tetradecanoylphorbol Acetate/pharmacology
2.
Pharmacol Ther ; 165: 14-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27179745

ABSTRACT

Smooth muscle cells provide crucial contractile functions in visceral, vascular, and lung tissues. The contractile state of smooth muscle is largely determined by their electrical excitability, which is in turn influenced by the activity of potassium channels. The activity of potassium channels sustains smooth muscle cell membrane hyperpolarization, reducing cellular excitability and thereby promoting smooth muscle relaxation. Research over the past decade has indicated an important role for Kv7 (KCNQ) voltage-gated potassium channels in the regulation of the excitability of smooth muscle cells. Expression of multiple Kv7 channel subtypes has been demonstrated in smooth muscle cells from viscera (gastrointestinal, bladder, myometrial), from the systemic and pulmonary vasculature, and from the airways of the lung, from multiple species, including humans. A number of clinically used drugs, some of which were developed to target Kv7 channels in other tissues, have been found to exert robust effects on smooth muscle Kv7 channels. Functional studies have indicated that Kv7 channel activators and inhibitors have the ability to relax and contact smooth muscle preparations, respectively, suggesting a wide range of novel applications for the pharmacological tool set. This review summarizes recent findings regarding the physiological functions of Kv7 channels in smooth muscle, and highlights potential therapeutic applications based on pharmacological targeting of smooth muscle Kv7 channels throughout the body.


Subject(s)
Drug Design , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth/drug effects , Potassium Channel Blockers/therapeutic use , Respiratory System/drug effects , Viscera/drug effects , Animals , Bronchoconstriction/drug effects , Bronchoconstrictor Agents/therapeutic use , Bronchodilator Agents/therapeutic use , Humans , KCNQ Potassium Channels/metabolism , Molecular Targeted Therapy , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Potassium Channel Blockers/adverse effects , Respiratory System/metabolism , Respiratory System/physiopathology , Signal Transduction/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Viscera/metabolism , Viscera/physiopathology
3.
Mol Pharmacol ; 86(3): 330-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24944189

ABSTRACT

Recent research suggests that smooth muscle cells express Kv7.4 and Kv7.5 voltage-activated potassium channels, which contribute to maintenance of their resting membrane voltage. New pharmacologic activators of Kv7 channels, ML213 (N-mesitybicyclo[2.2.1]heptane-2-carboxamide) and ICA-069673 N-(6-chloropyridin-3-yl)-3,4-difluorobenzamide), have been reported to discriminate among channels formed from different Kv7 subtypes. We compared the effects of ML213 and ICA-069673 on homomeric human Kv7.4, Kv7.5, and heteromeric Kv7.4/7.5 channels exogenously expressed in A7r5 vascular smooth muscle cells. We found that, despite its previous description as a selective activator of Kv7.2 and Kv7.4, ML213 significantly increased the maximum conductance of homomeric Kv7.4 and Kv7.5, as well as heteromeric Kv7.4/7.5 channels, and induced a negative shift of their activation curves. Current deactivation rates decreased in the presence of the ML213 (10 µM) for all three channel combinations. Mutants of Kv7.4 (W242L) and Kv7.5 (W235L), previously found to be insensitive to another Kv7 channel activator, retigabine, were also insensitive to ML213 (10 µM). In contrast to ML213, ICA-069673 robustly activated Kv7.4 channels but was significantly less effective on homomeric Kv7.5 channels. Heteromeric Kv7.4/7.5 channels displayed intermediate responses to ICA-069673. In each case, ICA-069673 induced a negative shift of the activation curves without significantly increasing maximal conductance. Current deactivation rates decreased in the presence of ICA-069673 in a subunit-specific manner. Kv7.4 W242L responded to ICA-069673-like wild-type Kv7.4, but a Kv7.4 F143A mutant was much less sensitive to ICA-069673. Based on these results, ML213 and ICA-069673 likely bind to different sites and are differentially selective among Kv7.4, Kv7.5, and Kv7.4/7.5 channel subtypes.


Subject(s)
Anilides/pharmacology , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , KCNQ Potassium Channels/agonists , Muscle, Smooth, Vascular/metabolism , Pyridines/pharmacology , Animals , Cells, Cultured , Humans , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/physiology , Muscle, Smooth, Vascular/cytology , Mutation , Patch-Clamp Techniques , Rats
4.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L476-86, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24441871

ABSTRACT

KCNQ (Kv7 family) potassium (K(+)) channels were recently found in airway smooth muscle cells (ASMCs) from rodent and human bronchioles. In the present study, we evaluated expression of KCNQ channels and their role in constriction/relaxation of rat airways. Real-time RT-PCR analysis revealed expression of KCNQ4 > KCNQ5 > KCNQ1 > KCNQ2 > KCNQ3, and patch-clamp electrophysiology detected KCNQ currents in rat ASMCs. In precision-cut lung slices, the KCNQ channel activator retigabine induced a concentration-dependent relaxation of small bronchioles preconstricted with methacholine (MeCh; EC50 = 3.6 ± 0.3 µM). Bronchoconstriction was also attenuated in the presence of two other structurally unrelated KCNQ channel activators: zinc pyrithione (ZnPyr; 1 µM; 22 ± 7%) and 2,5-dimethylcelecoxib (10 µM; 24 ± 8%). The same three KCNQ channel activators increased KCNQ currents in ASMCs by two- to threefold. The bronchorelaxant effects of retigabine and ZnPyr were prevented by inclusion of the KCNQ channel blocker XE991. A long-acting ß2-adrenergic receptor agonist, formoterol (10 nM), did not increase KCNQ current amplitude in ASMCs, but formoterol (1-1,000 nM) did induce a time- and concentration-dependent relaxation of rat airways, with a notable desensitization during a 30-min treatment or with repetitive treatments. Coadministration of retigabine (10 µM) with formoterol produced a greater peak and sustained reduction of MeCh-induced bronchoconstriction and reduced the apparent desensitization observed with formoterol alone. Our findings support a role for KCNQ K(+) channels in the regulation of airway diameter. A combination of a ß2-adrenergic receptor agonist with a KCNQ channel activator may improve bronchodilator therapy.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Bronchi/drug effects , Bronchoconstriction/drug effects , Bronchodilator Agents/pharmacology , KCNQ Potassium Channels/agonists , Acetylcholine/metabolism , Animals , Anthracenes/pharmacology , Asthma/drug therapy , Asthma/metabolism , Bronchoconstrictor Agents/pharmacology , Carbamates/pharmacology , Ethanolamines/pharmacology , Formoterol Fumarate , Keratolytic Agents/pharmacology , Male , Membrane Transport Modulators/pharmacology , Methacholine Chloride/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Organometallic Compounds/pharmacology , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...