Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Proc Natl Acad Sci U S A ; 119(34): e2202926119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969786

ABSTRACT

The Ca2+-activated SK4 K+ channel is gated by Ca2+-calmodulin (CaM) and is expressed in immune cells, brain, and heart. A cryoelectron microscopy (cryo-EM) structure of the human SK4 K+ channel recently revealed four CaM molecules per channel tetramer, where the apo CaM C-lobe and the holo CaM N-lobe interact with the proximal carboxyl terminus and the linker S4-S5, respectively, to gate the channel. Here, we show that phosphatidylinositol 4-5 bisphosphate (PIP2) potently activates SK4 channels by docking to the boundary of the CaM-binding domain. An allosteric blocker, BA6b9, was designed to act to the CaM-PIP2-binding domain, a previously untargeted region of SK4 channels, at the interface of the proximal carboxyl terminus and the linker S4-S5. Site-directed mutagenesis, molecular docking, and patch-clamp electrophysiology indicate that BA6b9 inhibits SK4 channels by interacting with two specific residues, Arg191 and His192 in the linker S4-S5, not conserved in SK1-SK3 subunits, thereby conferring selectivity and preventing the Ca2+-CaM N-lobe from properly interacting with the channel linker region. Immunohistochemistry of the SK4 channel protein in rat hearts showed a widespread expression in the sarcolemma of atrial myocytes, with a sarcomeric striated Z-band pattern, and a weaker occurrence in the ventricle but a marked incidence at the intercalated discs. BA6b9 significantly prolonged atrial and atrioventricular effective refractory periods in rat isolated hearts and reduced atrial fibrillation induction ex vivo. Our work suggests that inhibition of SK4 K+ channels by targeting drugs to the CaM-PIP2-binding domain provides a promising anti-arrhythmic therapy.


Subject(s)
Atrial Fibrillation , Calmodulin , Intermediate-Conductance Calcium-Activated Potassium Channels , Potassium Channel Blockers , Animals , Atrial Fibrillation/drug therapy , Calcium Signaling , Calmodulin/metabolism , Cryoelectron Microscopy , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Phosphatidylinositol 4,5-Diphosphate , Potassium Channel Blockers/pharmacology , Rats
2.
J Pharmacol Sci ; 148(1): 1-5, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924112

ABSTRACT

The KCa3.1 inhibition up-regulates IL-10 expression in regulatory T (Treg) cells in the recovery phase of inflammatory bowel disease (IBD) model mice; however, the underlying signaling pathway remains unclear. We investigated the involvement of AP-1 (Fos/Jun) and NF-κB in the expression of IL-10 and its transcription factors (TFs) in in vitro-induced mouse splenic Treg cells. The pharmacological inhibition of JNK reversed KCa3.1 inhibition-induced increases in the expression of IL-10 and its TFs. The inhibition of KCa3.1 increased phosphorylated JNK and c-Jun levels. Therefore, the JNK/c-Jun signaling pathway may contribute to the KCa3.1 inhibition-induced up-regulation of IL-10 in peripherally-induced Treg cells.


Subject(s)
Gene Expression/genetics , Inflammatory Bowel Diseases/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-jun/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Disease Models, Animal , Mice , NF-kappa B/metabolism , Phosphorylation , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Int J Mol Sci ; 22(19)2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34639190

ABSTRACT

Calcium signaling plays a vital role in the regulation of various cellular processes, including activation, proliferation, and differentiation of T-lymphocytes, which is mediated by ORAI1 and potassium (K+) channels. These channels have also been identified as highly attractive therapeutic targets for immune-related diseases. Licochalcone A is a licorice-derived chalconoid known for its multifaceted beneficial effects in pharmacological treatments, including its anti-inflammatory, anti-asthmatic, antioxidant, antimicrobial, and antitumorigenic properties. However, its anti-inflammatory effects involving ion channels in lymphocytes remain unclear. Thus, the present study aimed to investigate whether licochalcone A inhibits ORAI1 and K+ channels in T-lymphocytes. Our results indicated that licochalcone A suppressed all three channels (ORAI1, Kv1.3, and KCa3.1) in a concentration-dependent matter, with IC50 values of 2.97 ± 1.217 µM, 0.83 ± 1.222 µM, and 11.21 ± 1.07 µM, respectively. Of note, licochalcone A exerted its suppressive effects on the IL-2 secretion and proliferation in CD3 and CD28 antibody-induced T-cells. These results indicate that the use of licochalcone A may provide an effective treatment strategy for inflammation-related immune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Gene Expression Regulation/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , ORAI1 Protein/antagonists & inhibitors , T-Lymphocytes/drug effects , Calcium/metabolism , Calcium Signaling , HEK293 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Jurkat Cells , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
J Clin Lab Anal ; 35(8): e23854, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34313357

ABSTRACT

OBJECTIVE: In order to find new strategies for the prevention of vascular calcification in uremic individuals especially treated by dialysis and develop novel therapeutic targets in vascular calcification, we explore the role of KCa3.1 in alkalinization-induced VSMCs calcification in vitro. METHOD: Rat VSMCs calcification model was established by beta-glycerophosphate (ß-GP, 10 mM) induction. The pH of Dulbecco's modified Eagle's medium (DMEM) was adjusted every 24 h with 10 mM HCl or 10 mM NaHCO3 . The mineralization was measured by Alizarin Red staining and O-cresolphthalein complex one method. mRNA and protein expression were detected by RT-PCR and Western blot or immunofluorescence. Ca2+ influx was measured by Elisa. RESULT: The results indicated that alkalization induced an increase in Ca2+ influx to enhance VSMCs calcification. Furthermore, the increase of calcification was associated with the expression of KCa3.1 via advanced expression of osteoblastic differentiation markers alkaline phosphatase (ALP) and Runt-related transcription factor 2 (Runx2). Blocking KCa3.1 with TRAM-34 or shRNA vector can significantly lowered the effects of calcification in the activity of ALP and Runx2 expression. CONCLUSION: Together all, our studies suggested that alkalinization can promote vascular calcification by upregulating KCa3.1 channel and enhancing osteogenic/chondrogenic differentiation by upregulating Runx2. The specific inhibitor TRAM-34 and KCa3.1-shRNA ameliorated VSMCs calcification by downregulating KCa3.1.


Subject(s)
Aorta/pathology , Calcinosis/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Alkaline Phosphatase/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Calcinosis/chemically induced , Calcinosis/drug therapy , Calcium/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Glycerophosphates/toxicity , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pyrazoles/pharmacology , Rats, Sprague-Dawley
5.
Int J Mol Sci ; 22(13)2021 Jul 04.
Article in English | MEDLINE | ID: mdl-34281255

ABSTRACT

Midazolam (MDZ) could affect lymphocyte immune functions. However, the influence of MDZ on cell's K+ currents has never been investigated. Thus, in the present study, the effects of MDZ on Jurkat T lymphocytes were studied using the patch-clamp technique. Results showed that MDZ suppressed the amplitude of delayed-rectifier K+ current (IK(DR)) in concentration-, time-, and state-dependent manners. The IC50 for MDZ-mediated reduction of IK(DR) density was 5.87 µM. Increasing MDZ concentration raised the rate of current-density inactivation and its inhibitory action on IK(DR) density was estimated with a dissociation constant of 5.14 µM. In addition, the inactivation curve of IK(DR) associated with MDZ was shifted to a hyperpolarized potential with no change on the slope factor. MDZ-induced inhibition of IK(DR) was not reversed by flumazenil. In addition, the activity of intermediate-conductance Ca2+-activated K+ (IKCa) channels was suppressed by MDZ. Furthermore, inhibition by MDZ on both IK(DR) and IKCa-channel activity appeared to be independent from GABAA receptors and affected immune-regulating cytokine expression in LPS/PMA-treated human T lymphocytes. In conclusion, MDZ suppressed current density of IK(DR) in concentration-, time-, and state-dependent manners in Jurkat T-lymphocytes and affected immune-regulating cytokine expression in LPS/PMA-treated human T lymphocytes.


Subject(s)
Delayed Rectifier Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Midazolam/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Cytokines/metabolism , Delayed Rectifier Potassium Channels/metabolism , Dose-Response Relationship, Drug , Flumazenil/pharmacology , GABA-A Receptor Antagonists/pharmacology , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Jurkat Cells , Kinetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Microscopy, Confocal , Midazolam/administration & dosage , Patch-Clamp Techniques , Phytohemagglutinins/pharmacology , T-Lymphocytes/immunology
6.
Cell Calcium ; 96: 102384, 2021 06.
Article in English | MEDLINE | ID: mdl-33676318

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) metastases are the main cause of CRC mortality. Intracellular Ca2+ regulates cell migration and invasion, key factors for metastases. Ca2+ also activates Ca2+-dependent potassium channels which in turn affect Ca2+ driving force. We have previously reported that the expression of the Ca2+ activated potassium channel KCNN4 (SK4) is higher in CRC primary tumors compared to normal tissues. Here, we aimed to investigate the role of SK4 in the physiology of CRC. RESULTS: SK4 protein expression is enhanced in CRC tissues compared to normal colon tissues, with a higher level of KCNN4 in CRC patients with KRAS mutations. At the cellular level, we found that SK4 regulates the membrane potential of HCT116 cells. We also found that its inhibition reduced store operated Ca2+ entry (SOCE) and constitutive Ca2+ entry (CCE), while reducing cell migration. We also found that the activity of SK4 is linked to resistance pathways such as KRAS mutation and the expression of NRF2 and HIF-1α. In addition, the pharmacological inhibition of SK4 reduced intracellular reactive oxygen species (ROS) production, NRF2 expression and HIF1α stabilization. CONCLUSION: Our results suggest that SK4 contributes to colorectal cancer cell migration and invasion by modulating both Ca2+ entry and ROS regulation. Therefore, SK4 could be a potential target to reduce metastasis in KRAS-mutated CRC.


Subject(s)
Calcium/metabolism , Cell Movement/physiology , Colorectal Neoplasms/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Mutation/physiology , Proto-Oncogene Proteins p21(ras) , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Databases, Genetic , HCT116 Cells , HT29 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazoles/pharmacology
8.
BMC Pharmacol Toxicol ; 22(1): 6, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441172

ABSTRACT

BACKGROUND: Vigabatrin (VGB) is an approved non-traditional antiepileptic drug that has been revealed to have potential for treating brain tumors; however, its effect on ionic channels in glioma cells remains largely unclear. METHODS: With the aid of patch-clamp technology, we investigated the effects of VGB on various ionic currents in the glioblastoma multiforme cell line 13-06-MG. RESULTS: In cell-attached configuration, VGB concentration-dependently reduced the activity of intermediate-conductance Ca2+-activated K+ (IKCa) channels, while DCEBIO (5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one) counteracted the VGB-induced inhibition of IKCa channels. However, the activity of neither large-conductance Ca2+-activated (BKCa) nor inwardly rectifying K+ (KIR) channels were affected by the presence of VGB in human 13-06-MG cells. However, in the continued presence of VGB, the addition of GAL-021 or BaCl2 effectively suppressed BKCa and KIR channels. CONCLUSIONS: The inhibitory effect of VGB on IKCa channels demonstrated in the current study could be an important underlying mechanism of VGB-induced antineoplastic (e.g., anti-glioma) actions.


Subject(s)
Anticonvulsants/pharmacology , Antineoplastic Agents/pharmacology , Calcium Channel Blockers/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Vigabatrin/pharmacology , Brain Neoplasms/physiopathology , Cell Line, Tumor , Glioma/physiopathology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology
9.
J Pharmacol Exp Ther ; 377(1): 75-85, 2021 04.
Article in English | MEDLINE | ID: mdl-33504590

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract arising from abnormal responses of the innate and adaptative immune systems. Interleukin (IL)-10-producing CD4+CD25+ regulatory T (Treg) cells play a protective role in the recovery phase of IBD. In the present study, the effects of the administration of the selective Ca2+-activated K+ channel KCa3.1 inhibitor TRAM-34 on disease activities were examined in chemically induced IBD model mice. IBD disease severity, as assessed by diarrhea, visible fecal blood, inflammation, and crypt damage in the colon, was significantly lower in mice administered 1 mg/kg TRAM-34 than in vehicle-administered mice. Quantitative real-time polymerase chain reaction examinations showed that IL-10 expression levels in the recovery phase were markedly increased by the inhibition of KCa3.1 in mesenteric lymph node (mLN) Treg cells of IBD model mice compared with vehicle-administered mice. Among several positive and negative transcriptional regulators (TRs) for IL-10, three positive TRs-E4BP4, KLF4, and Blimp1-were upregulated by the inhibition of KCa3.1 in the mLN Treg cells of IBD model mice. In mouse peripheral CD4+CD25+ Treg cells induced by lectin stimulation, IL-10 expression and secretion were enhanced by the treatment with TRAM-34, together with the upregulation of E4BP4, KLF4, and Blimp1. Collectively, the present results demonstrated that the pharmacological inhibition of KCa3.1 decreased IBD symptoms in the IBD model by increasing IL-10 production in peripheral Treg cells and that IL-10high Treg cells produced by the treatment with KCa3.1 inhibitor may contribute to efficient Treg therapy for chronic inflammatory disorders, including IBD. SIGNIFICANCE STATEMENT: Pharmacological inhibition of Ca2+-activated K+ channel KCa3.1 increased IL-10 expression in peripheral Treg cells, together with the upregulation of the transcriptional regulators of IL-10: Krüppel-like factor 4, E4 promoter-binding protein 4, and/or B lymphocyte-induced maturation protein 1. The manipulation of IL-10high-producing Treg cells by the pharmacological inhibition of KCa3.1 may be beneficial in the treatment of chronic inflammatory diseases such as inflammatory bowel disease.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interleukin-10/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Membrane Transport Modulators/pharmacology , Pyrazoles/pharmacology , T-Lymphocytes, Regulatory/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Female , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , T-Lymphocytes, Regulatory/drug effects
10.
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
11.
ChemMedChem ; 15(24): 2462-2469, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33043595

ABSTRACT

The Ca2+ activated potassium channel 3.1 (KCa 3.1) is involved in critical steps of the metastatic cascade, such as proliferation, migration, invasion and extravasation. Therefore, a fast and efficient protocol for imaging of KCa 3.1 channels was envisaged. The novel fluorescently labeled small molecule imaging probes 1 and 2 were synthesized by connecting a dimethylpyrrole-based BODIPY dye with a derivative of the KCa 3.1 channel inhibitor senicapoc via linkers of different length. Patch-clamp experiments revealed the inhibition of KCa 3.1 channels by the probes confirming interaction with the channel. Both probes 1 and 2 were able to stain KCa 3.1 channels in non-small-cell lung cancer (NSCLC) cells following a simple, fast and efficient protocol. Pre-incubation with unlabeled senicapoc removed the punctate staining pattern showing the specificity of the new probes 1 and 2. Staining of the channel with the fluorescently labeled senicapoc derivatives 1 or 2 or with antibody-based indirect immunofluorescence yielded identical or very similar densities of stained KCa 3.1 channels. However, co-staining using both methods did not lead to the expected overlapping punctate staining pattern. This observation was explained by docking studies showing that the antibody used for indirect immunofluorescence and the probes 1 and 2 label different channel populations. Whereas the antibody binds at the closed channel conformation, the probes 1 and 2 bind within the open channel.


Subject(s)
Acetamides/pharmacology , Boron Compounds/pharmacology , Fluorescent Dyes/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Trityl Compounds/pharmacology , A549 Cells , Acetamides/metabolism , Animals , Antibodies/immunology , Antibodies/metabolism , Boron Compounds/metabolism , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Mice , Patch-Clamp Techniques , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Protein Binding , Staining and Labeling , Trityl Compounds/metabolism
12.
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
14.
PLoS One ; 15(3): e0222619, 2020.
Article in English | MEDLINE | ID: mdl-32150577

ABSTRACT

Ion channels have recently attracted attention as potential mediators of skin disease. Here, we explored the consequences of genetically encoded induction of the cell volume-regulating Ca2+-activated KCa3.1 channel (Kcnn4) for murine epidermal homeostasis. Doxycycline-treated mice harboring the KCa3.1+-transgene under the control of the reverse tetracycline-sensitive transactivator (rtTA) showed 800-fold channel overexpression above basal levels in the skin and solid KCa3.1-currents in keratinocytes. This overexpression resulted in epidermal spongiosis, progressive epidermal hyperplasia and hyperkeratosis, itch and ulcers. The condition was accompanied by production of the pro-proliferative and pro-inflammatory cytokines, IL-ß1 (60-fold), IL-6 (33-fold), and TNFα (26-fold) in the skin. Treatment of mice with the KCa3.1-selective blocker, Senicapoc, significantly suppressed spongiosis and hyperplasia, as well as induction of IL-ß1 (-88%) and IL-6 (-90%). In conclusion, KCa3.1-induction in the epidermis caused expression of pro-proliferative cytokines leading to spongiosis, hyperplasia and hyperkeratosis. This skin condition resembles pathological features of eczematous dermatitis and identifies KCa3.1 as a regulator of epidermal homeostasis and spongiosis, and as a potential therapeutic target.


Subject(s)
Eczema/genetics , Epidermis/pathology , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Keratosis/genetics , Skin/metabolism , Transgenes , Acetamides/pharmacology , Animals , Cytokines/metabolism , Doxycycline/pharmacology , Eczema/drug therapy , Female , Homeostasis/genetics , Hyperplasia/drug therapy , Hyperplasia/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Keratinocytes/metabolism , Keratosis/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Trans-Activators/metabolism , Trityl Compounds/pharmacology
15.
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
16.
Int J Pharm ; 580: 119180, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32135227

ABSTRACT

Senicapoc (SEN), a potent antisickling agent, shows poor water solubility and poor oral bioavailability. To improve the solubility and cell permeation of SEN, self-nanoemulsifying drug delivery systems (SNEDDSs) were developed. Capryol PGMC®, which showed the highest solubilization capacity, was selected as the oil. The self-emulsification ability of two surfactants, viz., Cremophor-EL® and Tween® 80, was compared. Based on a solubility study and ternary phase diagrams, three optimized nanoemulsions with droplet sizes less than 200 nm were prepared. An in vitro dissolution study demonstrated the superior performance of the SNEDDS over the free drug. During in vitro lipolysis, 80% of SEN loaded in the SNEDDS remained solubilized. An in vitro cytotoxicity study using the Caco-2 cell line indicated the safety of the formulations at 1 mg/mL. The transport of SEN-SNEDDSs across Caco-2 monolayers was enhanced 115-fold (p < 0.01) compared to that of the free drug. According to these results, SNEDDS formulations could be promising tools for the oral delivery of SEN.


Subject(s)
Acetamides/chemical synthesis , Drug Delivery Systems/methods , Drug Design , Emulsifying Agents/chemical synthesis , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Trityl Compounds/chemical synthesis , Acetamides/pharmacokinetics , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Emulsifying Agents/pharmacokinetics , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Solubility , Trityl Compounds/pharmacokinetics
17.
Heart Vessels ; 35(2): 284-289, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31473803

ABSTRACT

Flow-induced dilation in resistance arteries is mediated by endothelium-dependent hyperpolarisation via small and intermediate conducting Ca2+ sensitive K+ channels. The aim of the current study was to assess the effect of blocking both channels, using the toxins apamin and charybdotoxin, on flow-induced dilation in a conduit artery and vascular conductance. Experiments were carried out on the iliac and its vascular bed in anaesthetised pigs (n = 4). Flow-induced dilation and vascular conductance (∆F/∆P) were assessed before and after administration of toxins intra-arterially (i.a.) at 50 µg kg-1. Iliac diameter increased from baseline to 2.39 ± 0.4 mm before and 2.09 ± 0.46 mm after toxin administration, which was not significantly different (P = 0.63, Student's paired t test). Control conductance was 1.49 ± 0.27 ml min-1 mmHg-1 (P < 0.00001, ANOVA), and 1.53 ± 0.18 ml min-1 mmHg-1 (P < 0.00001, ANOVA) in the presence of the toxins which was not significantly different (P = 0.93 homogeneity of regression analysis). There was a small but significant increase in mean arterial pressure after the toxins were administered, from 74 ± 5 to 80 ± 9 mmHg (P = 0.03, Student's paired t test); but all other measured parameters were not significantly affected. Small- and intermediate-conducting Ca2+-sensitive K+ channels are not involved in flow-mediated dilation in conduit arteries and do not play a role in resistance vessel diameter maintenance at resting blood flow.


Subject(s)
Iliac Artery/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Vasodilation , Anesthesia, General , Animals , Arterial Pressure , Blood Flow Velocity , Female , Iliac Artery/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Models, Animal , Potassium Channel Blockers/pharmacology , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Sus scrofa , Vascular Resistance , Vasodilation/drug effects
18.
Sci Rep ; 9(1): 19893, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882807

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with limited therapeutic options and poor prognosis. IPF has been associated with aberrant vascular remodelling, however the role of vascular remodelling in pulmonary fibrosis is poorly understood. Here, we used a novel segmental challenge model of bleomycin-induced pulmonary fibrosis in sheep to evaluate the remodelling of the pulmonary vasculature, and to investigate the changes to this remodelling after the administration of the KCa3.1 channel inhibitor, senicapoc, compared to the FDA-approved drug pirfenidone. We demonstrate that in vehicle-treated sheep, bleomycin-infused lung segments had significantly higher blood vessel density when compared to saline-infused control segments in the same sheep. These microvascular density changes were significantly attenuated by senicapoc treatment. The increases in vascular endothelial growth factor (VEGF) expression and endothelial cell proliferation in bleomycin-infused lung segments were significantly reduced in sheep treated with the senicapoc, when compared to vehicle-treated controls. These parameters were not significantly suppressed with pirfenidone treatment. Senicapoc treatment attenuated vascular remodelling through inhibition of capillary endothelial cell proliferation and VEGF expression. These findings suggest a potential new mode of action for the novel drug senicapoc which may contribute to its efficacy in combatting pulmonary fibrosis.


Subject(s)
Bleomycin/adverse effects , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Lung/blood supply , Pulmonary Fibrosis/metabolism , Vascular Remodeling/drug effects , Acetamides/pharmacology , Animals , Bleomycin/pharmacology , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Lung/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Sheep , Trityl Compounds/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis
19.
Steroids ; 151: 108459, 2019 11.
Article in English | MEDLINE | ID: mdl-31330137

ABSTRACT

Basolateral membrane K+ channels play a key role in basal and agonist stimulated Cl- transport across airway epithelial cells by generating a favourable electrical driving force for Cl- efflux. The K+ channel sub-types and molecular mechanisms of regulation by hormones and secretagoues are still poorly understood. Here we have identified the type of K+ channels involved in cAMP and Ca2+ stimulated Cl- secretion and uncovered a novel anti-secretory effect of dexamethasone mediated by inhibition of basolateral membrane K+ channels in a human airway cell model of 16HBE14o- cells commonly used for ion transport studies. Dexamethasone produced a rapid inhibition of transepithelial chloride ion secretion under steady state conditions and after stimulation with cAMP agonist (forskolin) or a Ca2+ mobilizing agonist (ATP). Our results show three different types of K+ channels are targeted by dexamethasone to reduce airway secretion, namely Ca2+-activated secretion via KCNN4 (KCa3.1) channels and cAMP-activated secretion via KCNQ1 (Kv7.1) and KATP (Kir6.1,6.2) channels. The down-regulation of KCNN4 and KCNQ1 channel activities by dexamethasone involves rapid non-genomic activation of PKCα and PKA signalling pathways, respectively. Dexamethasone signal transduction for PKC and PKA activation was demonstrated to occur through a rapid non-genomic pathway that did not implicate the classical nuclear receptors for glucocorticoids or mineralocorticoids but occurred via a novel signalling cascade involving sequentially a Gi-protein coupled receptor, PKC, adenylyl cyclase Type IV, cAMP, PKA and ERK1/2 activation. The rapid, non-genomic, effects of dexamethasone on airway epithelial ion transport and cell signalling introduces a new paradigm for glucocorticoid actions in lung epithelia which may serve to augment the anti-inflammatory activity of the steroid and enhance its therapeutic potential in treating airway hypersecretion in asthma and COPD.


Subject(s)
Chlorides/metabolism , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , KATP Channels/antagonists & inhibitors , KCNQ1 Potassium Channel/antagonists & inhibitors , Bronchi/cytology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Gene Expression Regulation/drug effects , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , KATP Channels/metabolism , KCNQ1 Potassium Channel/metabolism , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Time Factors
20.
Ann Clin Transl Neurol ; 6(4): 723-738, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31019997

ABSTRACT

OBJECTIVE: Microglia play a pivotal role in the initiation and progression of Alzheimer's disease (AD). We here tested the therapeutic hypothesis that the Ca2+-activated potassium channel KCa3.1 constitutes a potential target for treating AD by reducing neuroinflammation. METHODS: To determine if KCa3.1 is relevant to AD, we tested if treating cultured microglia or hippocampal slices with Aß oligomer (AßO) activated KCa3.1 in microglia, and if microglial KCa3.1 was upregulated in 5xFAD mice and in human AD brains. The expression/activity of KCa3.1 was examined by qPCR, Western blotting, immunohistochemistry, and whole-cell patch-clamp. To investigate the role of KCa3.1 in AD pathology, we resynthesized senicapoc, a clinically tested KCa3.1 blocker, and determined its pharmacokinetic properties and its effect on microglial activation, Aß deposition and hippocampal long-term potentiation (hLTP) in 5xFAD mice. RESULTS: We found markedly enhanced microglial KCa3.1 expression/activity in brains of both 5xFAD mice and AD patients. In hippocampal slices, microglial KCa3.1 expression/activity was increased by AßO treatment, and its inhibition diminished the proinflammatory and hLTP-impairing activities of AßO. Senicapoc exhibited excellent brain penetrance and oral availability, and in 5xFAD mice, reduced neuroinflammation, decreased cerebral amyloid load, and enhanced hippocampal neuronal plasticity. INTERPRETATION: Our results prompt us to propose repurposing senicapoc for AD clinical trials, as senicapoc has excellent pharmacological properties and was safe and well-tolerated in a prior phase-3 clinical trial for sickle cell anemia. Such repurposing has the potential to expedite the urgently needed new drug discovery for AD.


Subject(s)
Acetamides/pharmacology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Trityl Compounds/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Drug Repositioning/methods , Humans , Mice, Transgenic , Microglia/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...