Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
JCI Insight ; 5(16)2020 08 20.
Article in English | MEDLINE | ID: mdl-32814712

ABSTRACT

Airway mucociliary clearance (MCC) is the main mechanism of lung defense keeping airways free of infection and mucus obstruction. Airway surface liquid volume, ciliary beating, and mucus are central for proper MCC and critically regulated by sodium absorption and anion secretion. Impaired MCC is a key feature of muco-obstructive diseases. The calcium-activated potassium channel KCa.3.1, encoded by Kcnn4, participates in ion secretion, and studies showed that its activation increases Na+ absorption in airway epithelia, suggesting that KCa3.1-induced hyperpolarization was sufficient to drive Na+ absorption. However, its role in airway epithelium is not fully understood. We aimed to elucidate the role of KCa3.1 in MCC using a genetically engineered mouse. KCa3.1 inhibition reduced Na+ absorption in mouse and human airway epithelium. Furthermore, the genetic deletion of Kcnn4 enhanced cilia beating frequency and MCC ex vivo and in vivo. Kcnn4 silencing in the Scnn1b-transgenic mouse (Scnn1btg/+), a model of muco-obstructive lung disease triggered by increased epithelial Na+ absorption, improved MCC, reduced Na+ absorption, and did not change the amount of mucus but did reduce mucus adhesion, neutrophil infiltration, and emphysema. Our data support that KCa3.1 inhibition attenuated muco-obstructive disease in the Scnn1btg/+ mice. K+ channel modulation may be a therapeutic strategy to treat muco-obstructive lung diseases.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Lung Diseases, Obstructive/etiology , Mucociliary Clearance/physiology , Animals , Calcium/metabolism , Cells, Cultured , Cilia/drug effects , Cilia/metabolism , Disease Models, Animal , Epithelium/metabolism , Female , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Lung/physiopathology , Lung Diseases, Obstructive/genetics , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Mucociliary Clearance/drug effects , Sodium/metabolism
2.
Nitric Oxide ; 99: 7-16, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32165314

ABSTRACT

Insulin regulates the l-arginine/nitric oxide (NO) pathway in human umbilical vein endothelial cells (HUVECs), increasing the plasma membrane expression of the l-arginine transporter hCAT-1 and inducing vasodilation in umbilical and placental veins. Placental vascular relaxation induced by insulin is dependent of large conductance calcium-activated potassium channels (BKCa), but the role of KCa channels on l-arginine transport and NO synthesis is still unknown. The aim of this study was to determine the contribution of KCa channels in both insulin-induced l-arginine transport and NO synthesis, and its relationship with placental vascular relaxation. HUVECs, human placental vein endothelial cells (HPVECs) and placental veins were freshly isolated from umbilical cords and placenta from normal pregnancies. Cells or tissue were incubated in absence or presence of insulin and/or tetraethylammonium, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole, iberiotoxin or NG-nitro-l-arginine methyl ester. l-Arginine uptake, plasma membrane polarity, NO levels, hCAT-1 expression and placenta vascular reactivity were analyzed. The inhibition of intermediate-conductance KCa (IKCa) and BKCa increases l-arginine uptake, which was related with protein abundance of hCAT-1 in HUVECs. IKCa and BKCa activities contribute to NO-synthesis induced by insulin but are not directly involved in insulin-stimulated l-arginine uptake. Long term incubation (8 h) with insulin increases the plasma membrane hyperpolarization and hCAT-1 expression in HUVECs and HPVECs. Insulin-induced relaxation in placental vasculature was reversed by KCa inhibition. The results show that the activity of IKCa and BKCa channels are relevant for both physiological regulations of NO synthesis and vascular tone regulation in the human placenta, acting as a part of negative feedback mechanism for autoregulation of l-arginine transport in HUVECs.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Nitric Oxide/metabolism , Umbilical Veins/metabolism , Adult , Arginine/metabolism , Cationic Amino Acid Transporter 1/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Insulin/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Peptides/pharmacology , Placenta/drug effects , Placenta/metabolism , Potassium Channel Blockers/pharmacology , Pregnancy , Pyrazoles/pharmacology , Umbilical Veins/drug effects , Young Adult
3.
Sci Rep ; 8(1): 9320, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29915289

ABSTRACT

Nearly 70% of cystic fibrosis (CF) patients bear the phenylalanine-508 deletion but disease severity differs greatly, and is not explained by the existence of different mutations in compound heterozygous. Studies demonstrated that genes other than CFTR relate to intestinal disease in humans and CF-mouse. Kcnn4, the gene encoding the calcium-activated potassium channel KCa3.1, important for intestinal secretion, is present in a locus linked with occurrence of intestinal CF-disease in mice and humans. We reasoned that it might be a CF-modifier gene and bred a CF-mouse with Kcnn4 silencing, finding that lethality was almost abolished. Silencing of Kcnn4 did not improve intestinal secretory functions, but rather corrected increased circulating TNF-α level and reduced intestinal mast cell increase. Given the importance of mast cells in intestinal disease additional double mutant CF-animals were tested, one lacking mast cells (C-kitW-sh/W-sh) and Stat6-/- to block IgE production. While mast cell depletion had no effect, silencing Stat6 significantly reduced lethality. Our results show that Kcnn4 is an intestinal CF modifier gene partially acting through a STAT6-dependent mechanism.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Genes, Modifier , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intestinal Diseases/genetics , Animals , Cytokines/metabolism , Immunoglobulin E/metabolism , Inflammation Mediators/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/deficiency , Intestinal Mucosa/pathology , Ion Channel Gating , Mast Cells/metabolism , Mice, Inbred C57BL , Mutation/genetics , Phenotype , STAT6 Transcription Factor/metabolism , Survival Analysis , Weight Gain
4.
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
5.
Acta Physiol (Oxf) ; 216(1): 132-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26138196

ABSTRACT

AIM: Neutrophils are the first cells to arrive at sites of injury. Nevertheless, many inflammatory diseases are characterized by an uncontrolled infiltration and action of these cells. Cell migration depends on volume changes that are governed by ion channel activity, but potassium channels in neutrophil have not been clearly identified. We aim to test whether KCa3.1 participates in neutrophil migration and other relevant functions of the cell. METHODS: Cytometer and confocal measurements to determine changes in cell volume were used. Cells isolated from human, mouse and horse were tested for KCa3.1-dependent chemotaxis. Chemokinetics, calcium handling and release of reactive oxygen species were measured to determine the role of KCa3.1 in those processes. A mouse model was used to test for neutrophil recruitment after acute lung injury in vivo. RESULTS: We show for the first time that KCa3.1 is expressed in mammalian neutrophils. When the channel is inhibited by a pharmacological blocker or by genetic silencing, it profoundly affects cell volume regulation, and chemotactic and chemokinetic properties of the cells. We also demonstrated that pharmacological inhibition of KCa3.1 did not affect calcium entry or reactive oxygen species production in neutrophils. Using a mouse model of acute lung injury, we observed that Kca3.1(-/-) mice are significantly less effective at recruiting neutrophils into the site of inflammation. CONCLUSIONS: These results demonstrate that KCa3.1 channels are key actors in the migration capacity of neutrophils, and its inhibition did not affect other relevant cellular functions.


Subject(s)
Calcium/metabolism , Chemotaxis , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neutrophils/metabolism , Animals , Humans , Inflammation , Membrane Potentials/physiology , Neutrophils/cytology
6.
PLoS One ; 10(9): e0139243, 2015.
Article in English | MEDLINE | ID: mdl-26418693

ABSTRACT

Formylated peptides are chemotactic agents generated by pathogens. The most relevant peptide is fMLF (formyl-Met-Leu-Phe) which participates in several immune functions, such as chemotaxis, phagocytosis, cytokine release and generation of reactive oxygen species. In macrophages fMLF-dependent responses are dependent on both, an increase in intracellular calcium concentration and on a hyperpolarization of the membrane potential. However, the molecular entity underlying this hyperpolarization remains unknown and it is not clear whether changes in membrane potential are linked to the increase in intracellular Ca2+. In this study, differentiated U937 cells, as a macrophage-like cell model, was used to characterize the fMLF response using electrophysiological and Ca2+ imaging techniques. We demonstrate by means of pharmacological and molecular biology tools that fMLF induces a Ca2+-dependent hyperpolarization via activation of the K+ channel KCa3.1 and thus, enhancing fMLF-induced intracellular Ca2+ increase through an amplification of the driving force for Ca2+ entry. Consequently, enhanced Ca2+ influx would in turn lengthen the hyperpolarization, operating as a positive feedback mechanism for fMLF-induced Ca2+ signaling.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Cell Differentiation/drug effects , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Feedback, Physiological/drug effects , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Monocytes/drug effects , Monocytes/metabolism , Monocytes/physiology , Patch-Clamp Techniques , RNA Interference , U937 Cells
7.
Free Radic Biol Med ; 52(5): 860-70, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22210378

ABSTRACT

Ca(2+)-activated K(+) channels (K(Ca)) and NO play a central role in the endothelium-dependent control of vasomotor tone. We evaluated the interaction of K(Ca) with NO production in isolated arterial mesenteric beds of the rat. In phenylephrine-contracted mesenteries, acetylcholine (ACh)-induced vasodilation was reduced by NO synthase (NOS) inhibition with N(ω)-nitro-L-arginine (L-NA), but in the presence of tetraethylammonium, L-NA did not further affect the response. In KCl-contracted mesenteries, the relaxation elicited by 100 nM ACh or 1 µM ionomycin was abolished by L-NA, tetraethylammonium, or simultaneous blockade of small-conductance K(Ca) (SK(Ca)) channels with apamin and intermediate-conductance K(Ca) (IK(Ca)) channels with triarylmethane-34 (TRAM-34). Apamin-TRAM-34 treatment also abolished 100 nM ACh-activated NO production, which was associated with an increase in superoxide formation. Endothelial cell Ca(2+) buffering with BAPTA elicited a similar increment in superoxide. Apamin-TRAM-34 treatment increased endothelial NOS phosphorylation at threonine 495 (P-eNOS(Thr495)). Blockade of NAD(P)H oxidase with apocynin or superoxide dismutation with PEG-SOD prevented the increment in superoxide and changes in P-eNOS(Thr495) observed during apamin and TRAM-34 application. Our results indicate that blockade of SK(Ca) and IK(Ca) activates NAD(P)H oxidase-dependent superoxide formation, which leads to inhibition of NO release through P-eNOS(Thr495). These findings disclose a novel mechanism involved in the control of NO production.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Small-Conductance Calcium-Activated Potassium Channels/physiology , Acetylcholine/pharmacology , Animals , Apamin/pharmacology , Calcium Ionophores/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Ionomycin/pharmacology , Male , Membrane Potentials/drug effects , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Superoxides/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
8.
J Physiol ; 583(Pt 2): 705-17, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17584847

ABSTRACT

Intestinal fluid secretion is driven by apical membrane, cystic fibrosis transmembrane conductance regulator (CFTR)-mediated efflux of Cl- that is concentrated in cells by basolateral Na(+)-K(+)-2Cl- cotransporters (NKCC1). An absolute requirement for Cl- efflux is the parallel activation of K(+) channels which maintain a membrane potential that sustains apical anion secretion. Both cAMP and Ca(2+) are intracellular signals for intestinal Cl- secretion. The K(+) channel involved in cAMP-dependent secretion has been identified as the KCNQ1-KCNE3 complex, but the identity of the K(+) channel driving Ca(2+)-activated Cl- secretion is controversial. We have now used a Kcnn4 null mouse to show that the intermediate conductance IK1 K(+) channel is necessary and sufficient to support Ca(2+)-dependent Cl- secretion in large and small intestine. Ussing chambers were used to monitor transepithelial potential, resistance and equivalent short-circuit current in colon and jejunum from control and Kcnn4 null mice. Na(+), K(+) and water content of stools was also measured. Distal colon and small intestinal epithelia from Kcnn4 null mice had normal cAMP-dependent Cl- secretory responses. In contrast, they completely lacked Cl- secretion in response to Ca(2+)-mobilizing agonists. Ca(2+)-activated electrogenic K(+) secretion was increased in colon epithelium of mice deficient in the IK1 channel. Na(+) and water content of stools was diminished in IK1-null animals. The use of Kcnn4 null mice has allowed us to demonstrate that IK1 K(+) channels are solely responsible for driving intestinal Ca(2+)-activated Cl- secretion. The absence of this channel leads to a marked reduction in water content in the stools, probably as a consequence of decreased electrolyte and water secretion.


Subject(s)
Body Water/metabolism , Calcium Signaling , Chlorides/metabolism , Colon/metabolism , Feces/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Intestinal Secretions/metabolism , Jejunum/metabolism , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Colon/drug effects , Cyclic AMP/metabolism , Diffusion Chambers, Culture , Electric Impedance , Glucose/metabolism , Histamine/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/deficiency , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intestinal Mucosa/metabolism , Jejunum/drug effects , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/pharmacology , Phenylalanine/metabolism , Potassium/metabolism , Potassium Channels, Calcium-Activated/metabolism , Sodium/metabolism , Time Factors
9.
J Comput Aided Mol Des ; 19(11): 771-89, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16374673

ABSTRACT

Selective inhibition of the intermediate-conductance Ca(2+)-activated K(+ )channel (IK (Ca)) by some clotrimazole analogs has been successfully modeled using topological charge indexes (TCI) and genetic neural networks (GNNs). A neural network monitoring scheme evidenced a highly non-linear dependence between the IK (Ca) blocking activity and TCI descriptors. Suitable subsets of descriptors were selected by means of genetic algorithm. Bayesian regularization was implemented in the network training function with the aim of assuring good generalization qualities to the predictors. GNNs were able to yield a reliable predictor that explained about 97% data variance with good predictive ability. On the contrary, the best multivariate linear equation with descriptors selected by linear genetic search, only explained about 60%. In spite of when using the descriptors from the linear equations to train neural networks yielded higher fitted models, such networks were very unstable and had relative low predictive ability. However, the best GNN BRANN 2 had a Q ( 2 ) of LOO of cross-validation equal to 0.901 and at the same time exhibited outstanding stability when calculating 80 randomly constructed training/test sets partitions. Our model suggested that structural fragments of size three and seven have relevant influence on the inhibitory potency of the studied IK (Ca) channel blockers. Furthermore, inhibitors were well distributed regarding its activity levels in a Kohonen self-organizing map (KSOM) built using the inputs of the best neural network predictor.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Methane/analogs & derivatives , Algorithms , Clotrimazole/analogs & derivatives , Clotrimazole/chemistry , Clotrimazole/pharmacology , Drug Design , Electrochemistry , Methane/chemistry , Methane/pharmacology , Models, Chemical , Neural Networks, Computer , Quantitative Structure-Activity Relationship , Software Design , Trityl Compounds/chemistry , Trityl Compounds/pharmacology
10.
Toxicon ; 46(4): 418-29, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16026809

ABSTRACT

From the venom of the Mexican scorpion Centruroides elegans Thorell five peptides were isolated to homogeneity by chromatographic procedures and their full amino acid sequence was determined by automatic Edman degradation. They all belong to the Noxiustoxin subfamily of scorpion toxins and were given the systematic names alpha-KTx 2.8 to 2.12, with trivial names Ce1 to Ce5, respectively. They have 39 amino acid residues, except for Ce3 which has only 38, but all of them have three disulfide bridges, and have molecular weights of 4255, 4267, 4249, 4295 and 4255 atomic mass units, respectively for Ce1 to Ce5. The C-terminal residues of Ce2, Ce4 and Ce5 were found to be amidated. The electrophysiological assay (whole-cell patch-clamp) showed that out of the five peptides, Ce1 (alpha-KTx 2.8), Ce2 (alpha-KTX2.9) and Ce4 (alpha-KTx 2.11) were effective blockers of Kv1.3 channels of human T lymphocytes, whereas these peptides did not inhibit the Ca2+-activated K+ channels (IKCa1) of the same cells. The equilibrium dissociation constants of these peptides for Kv1.3 were 0.70, 0.25 and 0.98nM for Ce1, Ce2 and Ce4, respectively. Furthermore, toxins Ce1, Ce2 and Ce4 practically did not inhibit the related voltage gated Shaker K+ channels, and rKv2.1 channels of the Shab family. The high affinity blockage of Kv1.3 channels by these peptides and their selectivity for Kv1.3 over IKCa1 may have significance in the development of novel tools for suppressing the function of those T cell subsets whose proliferation critically depends on the activity of Kv1.3 channels.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Peptides/genetics , Peptides/metabolism , Scorpion Venoms/chemistry , Scorpions , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Bayes Theorem , Cell Line , Chromatography, High Pressure Liquid , Electrophysiology , Enzyme-Linked Immunosorbent Assay , Humans , Mass Spectrometry , Mexico , Models, Genetic , Molecular Sequence Data , Organophosphorus Compounds , Peptides/toxicity , Phylogeny , Scorpion Venoms/toxicity , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL