Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Pulm Pharmacol Ther ; 23(5): 456-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20188205

ABSTRACT

Sildenafil, a phosphodiesterase-5 inhibitor, and simvastatin, a cholesterol lowering drug, both have therapeutic effects on PAH; however, the combination of these drugs has not been tested in the treatment of PAH. The purpose of this study was to determine whether the combination of sildenafil and simvastatin is superior to each drug alone in the prevention of MCT-induced PAH. Phosphorylated Smad levels were decreased in lung tissue in MCT-injected rats, whereas ERK protein levels were increased. This indicates a possible role for an increase in mitogenic ERK activity in addition to decreased proapoptotic Smad signaling in the MCT model of PAH. Combination sildenafil and simvastatin treatment prevented the MCT-induced increases in right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH), exerted an anti-proliferative effect on pulmonary artery smooth muscle cells (PASMC). Our results indicate that combination therapy with sildenafil and simvastatin attenuated the development of pulmonary hypertension more than either treatment alone.


Subject(s)
Anticholesteremic Agents/therapeutic use , Hypertension, Pulmonary/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Piperazines/therapeutic use , Simvastatin/therapeutic use , Sulfones/therapeutic use , Animals , Anticholesteremic Agents/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Male , Monocrotaline , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Purines/therapeutic use , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Simvastatin/pharmacology , Sulfones/pharmacology
3.
Am J Physiol Cell Physiol ; 298(3): C496-509, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20018952

ABSTRACT

The activity of voltage-gated K(+) (K(V)) channels plays an important role in regulating pulmonary artery smooth muscle cell (PASMC) contraction, proliferation, and apoptosis. The highly conserved NH(2)-terminal tetramerization domain (T1) of K(V) channels is important for proper channel assembly, association with regulatory K(V) beta-subunits, and localization of the channel to the plasma membrane. We recently reported two nonsynonymous mutations (G182R and E211D) in the KCNA5 gene of patients with idiopathic pulmonary arterial hypertension, which localize to the T1 domain of KCNA5. To study the electrophysiological properties and expression patterns of the mutants compared with the wild-type (WT) channel in vitro, we transfected HEK-293 cells with WT KCNA5, G182R, E211D, or the double mutant G182R/E211D channel. The mutants form functional channels; however, whole cell current kinetic differences between WT and mutant channels exist. Steady-state inactivation curves of the G182R and G182R/E211D channels reveal accelerated inactivation; the mutant channels inactivated at more hyperpolarized potentials compared with the WT channel. Channel protein expression was also decreased by the mutations. Compared with the WT channel, which was present in its mature glycosylated form, the mutant channels are present in greater proportion in their immature form in HEK-293 cells. Furthermore, G182R protein level is greatly reduced in COS-1 cells compared with WT. Immunostaining data support the hypothesis that, while WT protein localizes to the plasma membrane, mutant protein is mainly retained in intracellular packets. Overall, these data support a role for the T1 domain in channel kinetics as well as in KCNA5 channel subcellular localization.


Subject(s)
Kv1.5 Potassium Channel/metabolism , Potassium/metabolism , 4-Aminopyridine/pharmacology , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Glycosylation , Humans , Kinetics , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/chemistry , Kv1.5 Potassium Channel/genetics , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Mutation , Polymorphism, Single Nucleotide , Potassium Channel Blockers/pharmacology , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Transfection
4.
Ann N Y Acad Sci ; 1177: 101-11, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19845612

ABSTRACT

Acute hypoxia induces pulmonary vasoconstriction and chronic hypoxia causes pulmonary vascular remodeling characterized by significant vascular medial hypertrophy. Electromechanical and pharmacomechanical mechanisms are involved in regulating pulmonary vasomotor tone, while changes in cytosolic Ca2+ concentration ([Ca2+](cyt)) are an important signal in regulating contraction and proliferation of pulmonary artery smooth muscle cells (PASMC). Hypoxia-induced increases in [Ca2+](cyt) are, in part, mediated by selective inhibition of voltage-gated K+ (Kv) channels in PASMC. Kv1.5, encoded by the KCNA5 gene, is a Kv channel alpha subunit that forms functional homotetrameric and heterotetrameric Kv channels in PASMC. Activity of Kv channels contributes to the regulation of resting membrane potential. Overexpression of the human KCNA5 gene in rat PASMC and other cell types increases whole-cell Kv currents and causes membrane hyperpolarization. However, acute hypoxia only reduced Kv currents in KCNA5-transfected PASMC. These results provide compelling evidence that Kv1.5 is an important hypoxia-sensitive Kv channel in PASMC, contributing to regulation of membrane potential and intracellular Ca2+ homeostasis during hypoxia. This hypoxia-sensitive mechanism essential for inhibiting Kv1.5 channel activity is exclusively present in PASMC.


Subject(s)
Hypoxia/physiopathology , Kv1.5 Potassium Channel/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Kv1.5 Potassium Channel/genetics , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Rats
5.
Am J Physiol Cell Physiol ; 293(3): C928-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17581857

ABSTRACT

Mice are useful animal models to study pathogenic mechanisms involved in pulmonary vascular disease. Altered expression and function of voltage-gated K(+) (K(V)) channels in pulmonary artery smooth muscle cells (PASMCs) have been implicated in the development of pulmonary arterial hypertension. K(V) currents (I(K(V))) in mouse PASMCs have not been comprehensively characterized. The main focus of this study was to determine the biophysical and pharmacological properties of I(K(V)) in freshly dissociated mouse PASMCs with the patch-clamp technique. Three distinct whole cell I(K(V)) were identified based on the kinetics of activation and inactivation: rapidly activating and noninactivating currents (in 58% of the cells tested), rapidly activating and slowly inactivating currents (23%), and slowly activating and noninactivating currents (17%). Of the cells that demonstrated the rapidly activating noninactivating current, 69% showed I(K(V)) inhibition with 4-aminopyridine (4-AP), while 31% were unaffected. Whole cell I(K(V)) were very sensitive to tetraethylammonium (TEA), as 1 mM TEA decreased the current amplitude by 32% while it took 10 mM 4-AP to decrease I(K(V)) by a similar amount (37%). Contribution of Ca(2+)-activated K(+) (K(Ca)) channels to whole cell I(K(V)) was minimal, as neither pharmacological inhibition with charybdotoxin or iberiotoxin nor perfusion with Ca(2+)-free solution had an effect on the whole cell I(K(V)). Steady-state activation and inactivation curves revealed a window K(+) current between -40 and -10 mV with a peak at -31.5 mV. Single-channel recordings revealed large-, intermediate-, and small-amplitude currents, with an averaged slope conductance of 119.4 +/- 2.7, 79.8 +/- 2.8, 46.0 +/- 2.2, and 23.6 +/- 0.6 pS, respectively. These studies provide detailed electrophysiological and pharmacological profiles of the native K(V) currents in mouse PASMCs.


Subject(s)
Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Potassium Channels, Voltage-Gated/physiology , Pulmonary Artery/cytology , 4-Aminopyridine/pharmacology , Animals , Cells, Cultured , Charybdotoxin/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Muscle, Smooth, Vascular/cytology , Neurotoxins/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Tetraethylammonium/pharmacology
6.
Am J Physiol Cell Physiol ; 292(5): C1837-53, 2007 May.
Article in English | MEDLINE | ID: mdl-17267549

ABSTRACT

The pore-forming alpha-subunit, Kv1.5, forms functional voltage-gated K(+) (Kv) channels in human pulmonary artery smooth muscle cells (PASMC) and plays an important role in regulating membrane potential, vascular tone, and PASMC proliferation and apoptosis. Inhibited Kv channel expression and function have been implicated in PASMC from patients with idiopathic pulmonary arterial hypertension (IPAH). Here, we report that overexpression of the Kv1.5 channel gene (KCNA5) in human PASMC and other cell lines produced a 15-pS single channel current and a large whole cell current that was sensitive to 4-aminopyridine. Extracellular application of nicotine, bepridil, correolide, and endothelin-1 (ET-1) all significantly and reversibly reduced the Kv1.5 currents, while nicotine and bepridil also accelerated the inactivation kinetics of the currents. Furthermore, we sequenced KCNA5 from IPAH patients and identified 17 single-nucleotide polymorphisms (SNPs); 7 are novel SNPs. There are 12 SNPs in the upstream 5' region, 2 of which may alter transcription factor binding sites in the promoter, 2 nonsynonymous SNPs in the coding region, 2 SNPs in the 3'-untranslated region, and 1 SNP in the 3'-flanking region. Two SNPs may correlate with the nitric oxide-mediated decrease in pulmonary arterial pressure. Allele frequency of two other SNPs in patients with a history of fenfluramine and phentermine use was significantly different from patients who have never taken the anorexigens. These results suggest that 1) Kv1.5 channels are modulated by various agonists (e.g., nicotine and ET-1); 2) novel SNPs in KCNA5 are present in IPAH patients; and 3) SNPs in the promoter and translated regions of KCNA5 may underlie the altered expression and/or function of Kv1.5 channels in PASMC from IPAH patients.


Subject(s)
Hypertension, Pulmonary/genetics , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Myocytes, Smooth Muscle/metabolism , Polymorphism, Single Nucleotide , Pulmonary Artery/metabolism , Administration, Inhalation , Amino Acid Sequence , Animals , Antihypertensive Agents/administration & dosage , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Gene Frequency , Genotype , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Male , Membrane Potentials , Middle Aged , Molecular Sequence Data , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/administration & dosage , Patch-Clamp Techniques , Phenotype , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Transfection , Treatment Outcome
7.
Am J Physiol Cell Physiol ; 290(3): C907-16, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16236819

ABSTRACT

Acute hypoxia causes pulmonary vasoconstriction in part by inhibiting voltage-gated K(+) (Kv) channel activity in pulmonary artery smooth muscle cells (PASMC). The hypoxia-mediated decrease in Kv currents [I(K(V))] is selective to PASMC; hypoxia has little effect on I(K(V)) in mesenteric artery smooth muscle cells (MASMC). Functional Kv channels are homo- and/or heterotetramers of pore-forming alpha-subunits and regulatory beta-subunits. KCNA5 is a Kv channel alpha-subunit that forms functional Kv channels in PASMC and regulates resting membrane potential. We have shown that acute hypoxia selectively inhibits I(K(V)) through KCNA5 channels in PASMC. Overexpression of the human KCNA5 gene increased I(K(V)) and caused membrane hyperpolarization in HEK-293, COS-7, and rat MASMC and PASMC. Acute hypoxia did not affect I(K(V)) in KCNA5-transfected HEK-293 and COS-7 cells. However, overexpression of KCNA5 in PASMC conferred its sensitivity to hypoxia. Reduction of Po(2) from 145 to 35 mmHg reduced I(K(V)) by approximately 40% in rat PASMC transfected with human KCNA5 but had no effect on I(K(V)) in KCNA5-transfected rat MASMC (or HEK and COS cells). These results indicate that KCNA5 is an important Kv channel that regulates resting membrane potential and that acute hypoxia selectively reduces KCNA5 channel activity in PASMC relative to MASMC and other cell types. Because Kv channels (including KCNA5) are ubiquitously expressed in PASMC and MASMC, the observation from this study indicates that a hypoxia-sensitive mechanism essential for inhibiting KCNA5 channel activity is exclusively present in PASMC. The divergent effect of hypoxia on I(K(V)) in PASMC and MASMC also may be due to different expression levels of KCNA5 channels.


Subject(s)
Cell Hypoxia/physiology , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Animals , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Kv1.5 Potassium Channel/genetics , Male , Membrane Potentials , Mesenteric Arteries/cytology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...