Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Bol. latinoam. Caribe plantas med. aromát ; 21(4): 464-484, jul. 2022. tab, ilus, graf
Article in English | LILACS | ID: biblio-1526711

ABSTRACT

Abstract: The aim of this study was to evaluate the functional interaction of Glycyrrhiza glabra root extract (GGRE) on the large conductance Ca 2+ - activated K + (BKCa) channels expressed in the peripheral nervo us system by using nociception and inflammation models in rodents in vivo . Besides toxicity studies and open field tests, nociception and inflammation tests were performed on rodents. Different doses of GGRE were given orally to rats and mice. Naloxone, in domethacin, morphine, NS1619 and iberiotoxin (IbTX) were administered. GGRE had both anti - nociceptive and anti - inflammatory activity in rats and mice. GGRE exhibited an analgesic effect by decreasing the time - course of the pain threshold or reaction time i n some nociceptive tests. Furthermore, GGRE reduced level of pro - inflammatory cytokines, including TNF - α and IL - 1ß. As a conclusion, GGRE can alleviate the pain sensation of the afferent nerves and can reduce inflammation and associated pain by activating B KCa channels and reducing the levels of TNF - α, IL1ß


Resumen: El objetivo de este estudio fue evaluar la interacción funcional del extracto de raíz de Glycyrr hiza glabra (GGRE) en los canales de K + (BKCa) activados por Ca 2+ de gran conductancia expresados en el sistema nervioso periférico mediante el uso de modelos de nocicepción e inflamación en roedores in vivo . Además de los estudios de toxicidad y las prueb as de campo abierto, se realizaron pruebas de nocicepción e inflamación en roedores. Se administraron por vía oral diferentes dosis de GGRE a ratas y ratones. Se administraron naloxona, indometacina, morfina, NS1619 e iberiotoxina (IbTX). GGRE tenía activi dad tanto antinociceptiva como antiinflamatoria en ratas y ratones. GGRE mostró un efecto analgésico al disminuir la evolución temporal del umbral del dolor o el tiempo de reacción en algunas pruebas nociceptivas. Además, GGRE redujo el nivel de citocinas proinflamatorias, incluidas TNF - α e IL - 1ß. Como conclusión, GGRE puede aliviar la sensación de dolor de los nervios aferentes y puede reducir la inflamación y el dolor asociado activando los canales BKCa y reduciendo los niveles de TNF - α, IL1ß.


Subject(s)
Animals , Rats , Plant Extracts/administration & dosage , Glycyrrhiza/chemistry , Neuralgia/drug therapy , Phenols/analysis , Plant Extracts/pharmacology , Plant Extracts/chemistry , Analysis of Variance , Rats, Wistar , Plant Roots , Models, Animal , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Nociception/drug effects , Inflammation
2.
Cephalalgia ; 40(11): 1145-1154, 2020 10.
Article in English | MEDLINE | ID: mdl-32847403

ABSTRACT

INTRODUCTION: Preclinical data implicate large conductance calcium-activated potassium (BKCa) channels in the pathogenesis of headache and migraine, but the exact role of these channels is still unknown. Here, we investigated whether opening of BKCa channels would cause headache and vascular effects in healthy volunteers. METHODS: In a randomized, double-blind, placebo-controlled, cross-over study, 21 healthy volunteers aged 18-39 years were randomly allocated to receive an intravenous infusion of 0.05 mg/min BKCa channel opener MaxiPost and placebo on two different days. The primary endpoints were the difference in incidence of headache and the difference in area under the curve (AUC) for headache intensity scores (0-12 hours) and for middle cerebral artery blood flow velocity (VMCA) (0-2 hours) between MaxiPost and placebo. The secondary endpoints were the differences in area under the curve for superficial temporal artery and radial artery diameter (0-2 hours) between MaxiPost and placebo. RESULTS: Twenty participants completed the study. Eighteen participants (90%) developed headache after MaxiPost compared with six (30%) after placebo (p = 0.0005); the difference of incidence is 60% (95% confidence interval 36-84%). The area under the curve for headache intensity (AUC0-12 hours, p = 0.0003), for mean VMCA (AUC0-2 hours, p = 0.0001), for superficial temporal artery diameter (AUC0-2 hours, p = 0.003), and for radial artery diameter (AUC0-2 hours, p = 0.03) were significantly larger after MaxiPost compared to placebo. CONCLUSION: MaxiPost caused headache and dilation in extra- and intracerebral arteries. Our findings suggest a possible role of BKCa channels in headache pathophysiology in humans. ClinicalTrials.gov, ID: NCT03887325.


Subject(s)
Cerebrovascular Circulation/drug effects , Headache/metabolism , Hemodynamics/drug effects , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Adolescent , Adult , Cross-Over Studies , Double-Blind Method , Female , Headache/chemically induced , Healthy Volunteers , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Male , Vasodilator Agents/pharmacology , Young Adult
3.
Eur J Pharmacol ; 881: 173191, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32422186

ABSTRACT

Carbon monoxide (CO) is an endogenously synthesized gaseous mediator and is involved in the regulation of numerous physiological processes. Mitochondria, in which hemoproteins are abundant, are among the targets for CO action. Large-conductance calcium-activated (mitoBKCa) channels in the inner mitochondrial membrane share multiple biophysical similarities with the BKCa channels of the plasma membrane and could be a potential target for CO. To test this hypothesis, the activity of the mitoBKCa channels in human astrocytoma U-87 MG cell mitochondria was assessed with the patch-clamp technique. The effects of CO-releasing molecules (CORMs), such as CORM-2, CORM-401, and CORM-A1, were compared to the application of a CO-saturated solution to the mitoBKCa channels in membrane patches. The applied CORMs showed pleiotropic effects including channel inhibition, while the CO-containing solution did not significantly modulate channel activity. Interestingly, CO applied to the mitoBKCa channels, which were inhibited by exogenously added heme, stimulated the channel. To summarize, our findings indicate a requirement of heme binding to the mitoBKCa channel for channel modulation by CO and suggest that CORMs might have complex unspecific effects on mitoBKCa channels.


Subject(s)
Boranes/pharmacology , Carbon Monoxide/pharmacology , Carbonates/pharmacology , Heme/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Mitochondria/drug effects , N-substituted Glycines/pharmacology , Organometallic Compounds/pharmacology , Boranes/metabolism , Carbon Monoxide/metabolism , Carbonates/metabolism , Cell Line, Tumor , Heme/metabolism , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Potentials , Mitochondria/metabolism , N-substituted Glycines/metabolism , Organometallic Compounds/metabolism , Protein Binding
4.
Proc Natl Acad Sci U S A ; 117(2): 1021-1026, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31879339

ABSTRACT

The tremorgenic fungal alkaloid paxilline (PAX) is a commonly used specific inhibitor of the large-conductance, voltage- and Ca2+-dependent BK-type K+ channel. PAX inhibits BK channels by selective interaction with closed states. BK inhibition by PAX is best characterized by the idea that PAX gains access to the channel through the central cavity of the BK channel, and that only a single PAX molecule can interact with the BK channel at a time. The notion that PAX reaches its binding site via the central cavity and involves only a single PAX molecule would be consistent with binding on the axis of the permeation pathway, similar to classical open channel block and inconsistent with the observation that PAX selectively inhibits closed channels. To explore the potential sites of interaction of PAX with the BK channel, we undertook a computational analysis of the interaction of PAX with the BK channel pore gate domain guided by recently available liganded (open) and metal-free (closed) Aplysia BK channel structures. The analysis unambiguously identified a preferred position of PAX occupancy that accounts for all previously described features of PAX inhibition, including state dependence, G311 sensitivity, stoichiometry, and central cavity accessibility. This PAX-binding pose in closed BK channels is supported by additional functional results.


Subject(s)
Indoles/antagonists & inhibitors , Indoles/chemistry , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Animals , Binding Sites , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Mice , Molecular Docking Simulation , Protein Conformation , Protein Domains
5.
Pharmacol Rep ; 69(6): 1131-1139, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29128791

ABSTRACT

BACKGROUND: Some cannabinoids, a family of compounds derived from Cannabis sativa (marijuana), have previously shown vasodilator effects in several studies, a feature that makes them suitable for the generation of a potential treatment for hypertension. The mechanism underlying this vasodilator effect in arteries is still controversial. In this report, we explored how the synthetic cannabinoids ACPA (CB1-selective agonist) and JWH-133 (CB2-selective agonist) regulate the vascular tone of rat superior mesenteric arteries. METHODS: To screen the expression of CB1 (Cannabinoid receptor 1) and CB2 (Cannabinoid receptor 2) receptors in arterial rings or isolated smooth muscle cells obtained from the artery, immunocytochemistry, immunohistochemistry, and confocal microscopy were performed. In addition, the effects on vascular tone induced by the two cannabinoids were tested in isometric tension experiments in rings obtained from superior mesenteric arteries. The participation of voltage and calcium-activated potassium channel of big conductance (BKCa) and the role of nitric oxide (NO) release on the vascular effects induced by ACPA and JWH-133 were tested. RESULTS: CB1 and CB2 receptors were highly expressed in the rat superior mesenteric artery, in both smooth muscle and endothelium. The vasodilation effect shown by ACPA was endothelium-dependent through a mechanism involving CB1 receptors, BKCa channel activation, and NO release; meanwhile, the vasodilator effect of JWH-133 was induced by the activation of CB2 receptors located in smooth muscle and by a CB2 receptor-independent mechanism inducing NO release. CONCLUSIONS: CB1 and CB2 receptor activation in superior mesenteric artery causes vasorelaxation by mechanisms involving BKCa channels and NO release.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoids/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Male , Mesenteric Artery, Superior/drug effects , Mesenteric Artery, Superior/metabolism , Microscopy, Confocal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
6.
J Vasc Res ; 54(6): 329-343, 2017.
Article in English | MEDLINE | ID: mdl-29040972

ABSTRACT

AIM: The objective of this study was to examine the effects of n-3 polyunsaturated fatty acids (n-3 PUFAs) on coronary arterial large conductance Ca2+-activated K+ (BK) channel function in coronary smooth muscle cells (SMCs) of streptozotocin-induced diabetic rats. METHODS: The effects of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on coronary BK channel open probabilities were determined using the patch clamp technique. The mRNA and protein expressions of BK channel subunits were measured using qRT-PCR and Western blots. The coronary artery tension and coronary SMC Ca2+ concentrations were measured using a myograph system and fluorescence Ca2+ indicator. RESULTS: Compared to nondiabetic control rats, the BK channel function was impaired with a reduced response to EPA and DHA in freshly isolated SMCs of diabetic rats. Oral administration of n-3 PUFAs had no effects on protein expressions of BK channel subunits in nondiabetic rats, but significantly enhanced those of BK-ß1 in diabetic rats without altering BK-α protein levels. Moreover, coronary ring tension induced by iberiotoxin (a specific BK channel blocker) was increased and cytosolic Ca2+ concentrations in coronary SMCs were decreased in diabetic rats, but no changes were found in nondiabetic rats. CONCLUSIONS: n-3 PUFAs protect the coronary BK channel function and coronary vasoreactivity in diabetic rats as a result of not only increasing BK-ß1 protein expressions, but also decreasing coronary artery tension and coronary smooth muscle cytosolic Ca2+ concentrations.


Subject(s)
Coronary Artery Disease/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Angiopathies/prevention & control , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Vasoconstriction/drug effects , Animals , Calcium Signaling/drug effects , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Dose-Response Relationship, Drug , Drug Combinations , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Potassium Channel Blockers/pharmacology , Rats, Sprague-Dawley , Time Factors
7.
Handb Exp Pharmacol ; 240: 103-127, 2017.
Article in English | MEDLINE | ID: mdl-27838853

ABSTRACT

This chapter provides a critical overview of the available literature on the pharmacology of mitochondrial potassium channels. In the first part, the reader is introduced to the topic, and eight known protein contributors to the potassium permeability of the inner mitochondrial membrane are presented. The main part of this chapter describes the basic characteristics of each channel type mentioned in the introduction. However, the most important and valuable information included in this chapter concerns the pharmacology of mitochondrial potassium channels. Several available channel modulators are critically evaluated and rated by suitability for research use. The last figure of this chapter shows the results of this evaluation at a glance. Thus, this chapter can be very useful for beginners in this field. It is intended to be a time- and resource-saving guide for those searching for proper modulators of mitochondrial potassium channels.


Subject(s)
Mitochondria/metabolism , Potassium Channels/drug effects , Animals , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Potassium Channels/physiology , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/physiology
8.
PLoS Negl Trop Dis ; 9(10): e0004062, 2015.
Article in English | MEDLINE | ID: mdl-26437177

ABSTRACT

The anthelmintic emodepside paralyses adult filarial worms, via a mode of action distinct from previous anthelmintics and has recently garnered interest as a new treatment for onchocerciasis. Whole organism data suggest its anthelmintic action is underpinned by a selective activation of the nematode isoform of an evolutionary conserved Ca2+-activated K+ channel, SLO-1. To test this at the molecular level we compared the actions of emodepside at heterologously expressed SLO-1 alpha subunit orthologues from nematode (Caenorhabditis elegans), Drosophila melanogaster and human using whole cell voltage clamp. Intriguingly we found that emodepside modulated nematode (Ce slo-1), insect (Drosophila, Dm slo) and human (hum kcnma1)SLO channels but that there are discrete differences in the features of the modulation that are consistent with its anthelmintic efficacy. Nematode SLO-1 currents required 100 µM intracellular Ca2+ and were strongly facilitated by emodepside (100 nM; +73.0 ± 17.4%; n = 9; p < 0.001). Drosophila Slo currents on the other hand were activated by emodepside (10 µM) in the presence of 52 nM Ca2+ but were inhibited in the presence of 290 nM Ca2+ and exhibited a characteristic loss of rectification. Human Slo required 300 nM Ca2+ and emodepside transiently facilitated currents (100 nM; +33.5 ± 9%; n = 8; p<0.05) followed by a sustained inhibition (-52.6 ± 9.8%; n = 8; p < 0.001). This first cross phyla comparison of the actions of emodepside at nematode, insect and human channels provides new mechanistic insight into the compound's complex modulation of SLO channels. Consistent with whole organism behavioural studies on C. elegans, it indicates its anthelmintic action derives from a strong activation of SLO current, not observed in the human channel. These data provide an important benchmark for the wider deployment of emodepside as an anthelmintic treatment.


Subject(s)
Anthelmintics/pharmacology , Depsipeptides/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Drosophila melanogaster , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Protein Subunits
9.
Eur J Pharmacol ; 758: 177-87, 2015 Jul 05.
Article in English | MEDLINE | ID: mdl-25843414

ABSTRACT

Arecoline (ARE) is an alkaloid-type natural product from areca nut. This compound has numerous pharmacological and toxicological effects. Whether this agent interacts with ion channels to perturb functional activity of cells remains unknown. The effects of ARE on ionic currents were studied in glioma cell lines (U373 and U87MG) using patch-clamp technique. Like TRAM-34(1-[(2-chlorophenyl)-diphenylmethyl]pyrazole), ARE suppressed the amplitude of whole-cell voltage-gated K(+) currents in U373 cells elicited by a ramp voltage clamp. In cell-attached configuration, ARE did not modify the single-channel conductance of intermediate-conductance Ca(2+)-activated K(+) (IKCa) channels; however, it did reduce channel activity. Its inhibition of IKCa channels was accompanied by a significant lengthening in the slow component of mean closed time of IKCa channels. Based on minimal kinetic scheme, the dissociation constant (KD) required for ARE-mediated prolongation of mean closed time was 11.2µM. ARE-induced inhibition of IKCa channels was voltage-dependent. Inability of ARE to perturb the activity of large-conductance Ca(2+)-activated K(+) (BKCa) channels was seen. Under current-clamp recordings, ARE depolarized the membrane of U373 cells and DCEBIO reversed ARE-induced depolarization. Similarly, ARE suppressed IKCa-channel activities in oral keratinocytes. This study provides the evidence that ARE block IKCa channels in a concentration, voltage and state-dependent manner. ARE-induced block of IKCa channels is unrelated to the binding of muscarinic receptors. The effects of ARE on these channels may partially be responsible for the underlying cellular mechanisms by which it influences the functional activities of glioma cells or oral keratinocytes, if similar findings occur in vivo.


Subject(s)
Arecoline/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Arecoline/antagonists & inhibitors , Benzimidazoles/pharmacology , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Membrane Potentials/drug effects , Primary Cell Culture , Pyrazoles/pharmacology
10.
Physiol Res ; 64(1): 39-49, 2015.
Article in English | MEDLINE | ID: mdl-25194131

ABSTRACT

To investigate lisinopril effect on the contribution of nitric oxide (NO) and K(Ca) channels to acetylcholine (ACh)-induced relaxation in isolated mesenteric arteries of spontaneously hypertensive rats (SHRs). Third branch mesenteric arteries isolated from lisinopril treated SHR rats (20 mg/kg/day for ten weeks, SHR-T) or untreated (SHR-UT) or normotensive WKY rats were mounted on tension myograph and ACh concentration-response curves were obtained. Westernblotting of eNOS and K(Ca) channels was performed. ACh-induced relaxations were similar in all groups while L-NMMA and indomethacin caused significant rightward shift only in SHR-T group. Apamin and TRAM-34 (SK(Ca) and IK(Ca) channels blockers, respectively) significantly attenuated ACh-induced maximal relaxation by similar magnitude in vessels from all three groups. In the presence of L-NMMA, indomethacin, apamin and TRAM-34 further attenuated ACh-induced relaxation only in SHR-T. Furthermore, lisinopril treatment increased expression of eNOS, SK(Ca) and BK(Ca) proteins. Lisinopril treatment increased expression of eNOS, SK(Ca), BK(Ca) channel proteins and increased the contribution of NO to ACh-mediated relaxation. This increased role of NO was apparent only when EDHF component was blocked by inhibiting SK(Ca) and IK(Ca) channels. Such may suggest that in mesenteric arteries, non-EDHF component functions act as a reserve system to provide compensatory vasodilatation if (and when) hyperpolarization that is mediated by SK(Ca) and IK(Ca) channels is reduced.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Lisinopril/pharmacology , Mesenteric Arteries/drug effects , Nitric Oxide/metabolism , Potassium Channels, Calcium-Activated/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension/metabolism , Hypertension/physiopathology , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism
11.
Arthritis Rheumatol ; 67(1): 96-106, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25252152

ABSTRACT

OBJECTIVE: Fibroblast-like synoviocytes (FLS) participate in joint inflammation and damage in rheumatoid arthritis (RA) and its animal models. The purpose of this study was to define the importance of KCa1.1 (BK, Maxi-K, Slo1, KCNMA1) channel expression and function in FLS and to establish these channels as potential new targets for RA therapy. METHODS: We compared KCa1.1 expression levels in FLS from rats with pristane-induced arthritis (PIA) and in FLS from healthy rats. We then used ex vivo functional assays combined with small interfering RNA-induced knockdown, overexpression, and functional modulation of KCa1.1 in PIA FLS. Finally, we determined the effectiveness of modulating KCa1.1 in 2 rat models of RA, moderate PIA and severe collagen-induced arthritis (CIA). RESULTS: We found that PIA FLS expressed the KCa1.1 channel as their major potassium channel, as has been found in FLS from patients with RA. In contrast, FLS from healthy rats expressed fewer of these channels. Inhibiting the function or expression of KCa1.1 ex vivo reduced proliferation and invasive properties of, as well as protease production by, PIA FLS, whereas opening native KCa1.1 or overexpressing the channel enhanced the invasiveness of both FLS from rats with PIA and FLS from healthy rats. Treatment with a KCa1.1 channel blocker at the onset of clinical signs stopped disease progression in the PIA and CIA models, reduced joint and bone damage, and inhibited FLS invasiveness and proliferation. CONCLUSION: Our results demonstrate a critical role of KCa1.1 channels in the regulation of FLS invasiveness and suggest that KCa1.1 channels represent potential therapeutic targets in RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/prevention & control , Cell Movement/physiology , Fibroblasts/pathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Synovial Membrane/pathology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/chemically induced , Cell Proliferation/physiology , Disease Models, Animal , Female , Fibroblasts/physiology , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Matrix Metalloproteinase 2/physiology , Potassium Channel Blockers/pharmacology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Synovial Membrane/physiopathology , Terpenes/adverse effects
12.
Hypertension ; 65(2): 345-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25385762

ABSTRACT

Myogenic responses by resistance vessels are a key component of autoregulation in brain, thus playing a crucial role in regulating cerebral blood flow and protecting the blood-brain barrier against potentially detrimental elevations in blood pressure. Although cerebrovascular disease is often accompanied by alterations in myogenic responses, mechanisms that control these changes are poorly understood. Peroxisome proliferator-activated receptor γ has emerged as a regulator of vascular tone. We hypothesized that interference with peroxisome proliferator-activated receptor γ in smooth muscle would augment myogenic responses in cerebral arteries. We studied transgenic mice expressing a dominant-negative mutation in peroxisome proliferator-activated receptor γ selectively in smooth muscle (S-P467L) and nontransgenic littermates. Myogenic tone in middle cerebral arteries from S-P467L was elevated 3-fold when compared with nontransgenic littermates. Rho kinase is thought to play a major role in cerebrovascular disease. The Rho kinase inhibitor, Y-27632, abolished augmented myogenic tone in middle cerebral arteries from S-P467L mice. CN-03, which modifies RhoA making it constitutively active, elevated myogenic tone to ≈60% in both strains, via a Y-27632-dependent mechanism. Large conductance Ca(2+)-activated K(+) channels (BKCa) modulate myogenic tone. Inhibitors of BKCa caused greater constriction in middle cerebral arteries from nontransgenic littermates when compared with S-P467L. Expression of RhoA or Rho kinase-I/II protein was similar in cerebral arteries from S-P467L mice. Overall, the data suggest that peroxisome proliferator-activated receptor γ in smooth muscle normally inhibits Rho kinase and promotes BKCa function, thus influencing myogenic tone in resistance arteries in brain. These findings have implications for mechanisms that underlie large- and small-vessel disease in brain, as well as regulation of cerebral blood flow.


Subject(s)
Cerebrovascular Circulation/physiology , Middle Cerebral Artery/physiology , Muscle, Smooth, Vascular/physiology , PPAR gamma/deficiency , Vasoconstriction/physiology , Animals , Cerebrovascular Circulation/drug effects , Desoxycorticosterone Acetate/toxicity , Enzyme Induction , Gene Expression Profiling , Genes, Dominant , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Mice , Mice, Knockout , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/enzymology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , PPAR gamma/drug effects , PPAR gamma/genetics , PPAR gamma/physiology , Sodium Chloride/toxicity , Tetraethylammonium/pharmacology , rho GTP-Binding Proteins/biosynthesis , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/biosynthesis , rho-Associated Kinases/genetics , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein
13.
Ann Thorac Surg ; 99(2): 626-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25527424

ABSTRACT

BACKGROUND: Activation of large conductance calcium-activated potassium (BKCa) channels is cardioprotective for ischemic injury and can enhance vasorelaxation. Rottlerin has recently been identified as a potent BKCa activator. We demonstrated that rottlerin improves cardiac function and increases coronary flow when used as a cardioplegia additive in rat and mouse models of cardioplegic arrest and reperfusion. In this study we examined the effectiveness and specificity of the putative BKCa activator rottlerin on vascular reactivity in response to specific contractile and dilatory agonists. METHODS: Aortic rings from wild-type (wt) and BKCa knock-out (KO) mice were mounted in a tissue bath with force transducers. The vasodilatory effect of rottlerin was evaluated after pre-constriction with U46619. Dose responses to the contractile agonists U46619 and phenylephrine (PE), and vasodilation responses to rottlerin, hydrogen sulfide (H2S), and sodium nitroprusside (SNP) were performed after pretreatment with rottlerin. Similar studies were performed in pig coronary vessels. RESULTS: The BKCa KO mouse aortic rings exhibited spontaneous contraction and had greater contractile responses to U46619 and reduced vasodilation to SNP compared with wt mice. The wt and KO responses to phenylephrine were similar. Rottlerin dose dependently dilated wild-type vessels, but not in BKCa KO animals. Pretreatment with rottlerin caused depressed U46619 responses, but had no effect on PE, SNP, or H2S-mediated responses. However, pig coronary vessels pretreated with rottlerin exhibited reduced contractile responses and enhanced nitric oxide-dependent dilation. CONCLUSIONS: Rottlerin directly causes vasodilation through BKCa channel dependent mechanisms. The BKCa channel activator pretreatment enhances vasodilatory responses and impairs specific vasoconstrictive agonists.


Subject(s)
Acetophenones/pharmacology , Aorta/drug effects , Aorta/physiopathology , Benzopyrans/pharmacology , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Muscle Contraction/drug effects , Vasodilation/drug effects , Animals , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Knockout , Swine
14.
Elife ; 2: e01009, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24137539

ABSTRACT

Mammalian spermatozoa gain competence to fertilize an oocyte as they travel through the female reproductive tract. This process is accompanied by an elevation of sperm intracellular calcium and a membrane hyperpolarization. The latter is evoked by K(+) efflux; however, the molecular identity of the potassium channel of human spermatozoa (hKSper) is unknown. Here, we characterize hKSper, reporting that it is regulated by intracellular calcium but is insensitive to intracellular alkalinization. We also show that human KSper is inhibited by charybdotoxin, iberiotoxin, and paxilline, while mouse KSper is insensitive to these compounds. Such unique properties suggest that the Slo1 ion channel is the molecular determinant for hKSper. We show that Slo1 is localized to the sperm flagellum and is inhibited by progesterone. Inhibition of hKSper by progesterone may depolarize the spermatozoon to open the calcium channel CatSper, thus raising [Ca(2+)] to produce hyperactivation and allowing sperm to fertilize an oocyte. DOI:http://dx.doi.org/10.7554/eLife.01009.001.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Spermatozoa/metabolism , Calcium/metabolism , Charybdotoxin/pharmacology , Humans , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Male , Peptides/pharmacology , Progesterone/pharmacology , Sperm Tail/metabolism , Spermatozoa/drug effects
15.
Am J Physiol Renal Physiol ; 303(9): F1300-6, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22896041

ABSTRACT

Pharmacological blockade of cyclic nucleotide phosphodiesterase (PDE) can relax human urinary bladder smooth muscle (UBSM); however, the underlying cellular mechanism is unknown. In this study, we investigated the effects of PDE pharmacological blockade on human UBSM excitability, spontaneous and nerve-evoked contractility, and determined the underlying cellular mechanism mediating these effects. Patch-clamp electrophysiological experiments showed that 3-isobutyl-1-methylxanthine (10 µM), a nonselective PDE inhibitor, caused ∼3.6-fold increase in the transient K(Ca)1.1 channel current frequency and ∼2.5-fold increase in the spontaneous transient hyperpolarization frequency in UBSM-isolated cells. PDE blockade also caused ∼5.6-mV hyperpolarization of the UBSM cell membrane potential. Blocking the K(Ca)1.1 channels with paxilline abolished the spontaneous transient hyperpolarization and the hyperpolarization effect of PDE blockade on the UBSM cell membrane potential. Live cell Ca(2+)-imaging experiments showed that PDE blockade significantly decreased the global intracellular Ca(2+) levels. Attenuation of PDE activity significantly reduced spontaneous phasic contraction amplitude, muscle force integral, duration, frequency, and muscle tone of human UBSM isolated strips. Blockade of PDE also significantly reduced the contraction amplitude, muscle force integral, and duration of the nerve-evoked contractions induced by 20-Hz electrical field stimulation. Pharmacological inhibition of K(Ca)1.1 channels abolished the relaxation effects of PDE blockade on both spontaneous and nerve-evoked contractions in human UBSM-isolated strips. Our data provide strong evidence that in human UBSM PDE is constitutively active, thus maintaining spontaneous UBSM contractility. PDE blockade causes relaxation of human UBSM by increasing transient K(Ca)1.1 channel current activity, hyperpolarizing cell membrane potential, and decreasing the global intracellular Ca(2+).


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Muscle, Smooth/physiology , Phosphoric Diester Hydrolases/physiology , Urinary Bladder/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Aged , Female , Humans , In Vitro Techniques , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Middle Aged , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Potassium Channel Blockers/pharmacology , Urinary Bladder/drug effects
16.
Am J Physiol Heart Circ Physiol ; 303(3): H332-40, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22636679

ABSTRACT

We reported previously that tempol attenuated the exercise pressor and muscle mechanoreceptor reflexes in rats whose femoral arteries were ligated, whereas tempol did not attenuate these reflexes in rats whose femoral arteries were freely perfused. Although the mechanism whereby tempol attenuated these reflexes in rats whose femoral artery was ligated was independent of its ability to scavenge reactive oxygen species, its nature remains unclear. An alternative explanation for the tempol-induced attenuation of these reflexes involves ATP-sensitive potassium channels (K(ATP)) and calcium-activated potassium channels (BK(Ca)), both of which are opened by tempol. We tested the likelihood of this explanation by measuring the effects of either glibenclamide (0.1 mg/kg), which blocks K(ATP) channels, or iberiotoxin (20 or 40 µg/kg), which blocks BK(Ca) channels, on the tempol-induced attenuation of the exercise pressor and muscle mechanoreceptor reflexes in decerebrated rats whose femoral arteries were ligated. We found that glibenclamide prevented the tempol-induced attenuation of both reflexes, whereas iberiotoxin did not. We also found that the amount of protein comprising the pore of the K(ATP) channel in the dorsal root ganglia innervating hindlimbs whose femoral artery was ligated was significantly greater than that in the dorsal root ganglia innervating hindlimbs whose femoral arteries were freely perfused. In contrast, the amounts of protein comprising the BK(Ca) channel in the dorsal root ganglia innervating the ligated and freely perfused hindlimbs were not different. We conclude that tempol attenuated both reflexes by opening K(ATP) channels, an effect that hyperpolarized muscle afferents stimulated by static contraction or tendon stretch.


Subject(s)
Cyclic N-Oxides/pharmacology , Femoral Artery/surgery , Ganglia, Spinal/drug effects , Glyburide/pharmacology , KATP Channels/drug effects , Muscle Contraction , Muscle, Skeletal/blood supply , Muscle, Skeletal/innervation , Potassium Channel Blockers/pharmacology , Reflex/drug effects , Animals , Blood Pressure/drug effects , Blotting, Western , Cyclic N-Oxides/administration & dosage , Decerebrate State , Ganglia, Spinal/metabolism , Heart Rate/drug effects , Injections, Intra-Arterial , KATP Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Ligation , Male , Peptides/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Spin Labels , Time Factors
17.
Proc Natl Acad Sci U S A ; 109(9): 3552-7, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331907

ABSTRACT

Large-conductance voltage- and Ca(2+)-activated K(+) (Slo1 BK) channels serve numerous cellular functions, and their dysregulation is implicated in various diseases. Drugs activating BK channels therefore bear substantial therapeutic potential, but their deployment has been hindered in part because the mode of action remains obscure. Here we provide mechanistic insight into how the dehydroabietic acid derivative Cym04 activates BK channels. As a representative of NS1619-like BK openers, Cym04 reversibly left-shifts the half-activation voltage of Slo1 BK channels. Using an established allosteric BK gating model, the Cym04 effect can be simulated by a shift of the voltage sensor and the ion conduction gate equilibria toward the activated and open state, respectively. BK activation by Cym04 occurs in a splice variant-specific manner; it does not occur in such Slo1 BK channels using an alternative neuronal exon 9, which codes for the linker connecting the transmembrane segment S6 and the cytosolic RCK1 domain--the S6/RCK linker. In addition, Cym04 does not affect Slo1 BK channels with a two-residue deletion within this linker. Mutagenesis and model-based gating analysis revealed that BK openers, such as Cym04 and NS1619 but not mallotoxin, activate BK channels by functionally interacting with the S6/RCK linker, mimicking site-specific shortening of this purported passive spring, which transmits force from the cytosolic gating ring structure to open the channel's gate.


Subject(s)
Abietanes/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Potassium/metabolism , Allosteric Regulation , Amino Acid Sequence , HEK293 Cells , Humans , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism
18.
Pflugers Arch ; 461(6): 665-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21301863

ABSTRACT

The large conductance, voltage- and calcium-activated potassium channel, BK(Ca), is a known target for the gasotransmitter, carbon monoxide (CO). Activation of BK(Ca) by CO modulates cellular excitability and contributes to the physiology of a diverse array of processes, including vascular tone and oxygen-sensing. Currently, there is no consensus regarding the molecular mechanisms underpinning reception of CO by the BK(Ca). Here, employing voltage-clamped, inside-out patches from HEK293 cells expressing single, double and triple cysteine mutations in the BK(Ca) α-subunit, we test the hypothesis that CO regulation is conferred upon the channel by interactions with cysteine residues within the RCK2 domain. In physiological [Ca(2+)](i), all mutants carrying a cysteine substitution at position 911 (C911G) demonstrated significantly reduced CO sensitivity; the C911G mutant did not express altered Ca(2+)-sensitivity. In contrast, histidine residues in RCK1 domain, previously shown to ablate CO activation in low [Ca(2+)](i), actually increased CO sensitivity when [Ca(2+)](i) was in the physiological range. Importantly, cyanide, employed here as a substituent for CO at potential metal centres, occluded activation by CO; this effect was freely reversible. Taken together, these data suggest that a specific cysteine residue in the C-terminal domain, which is close to the Ca(2+) bowl but which is not involved in Ca(2+) activation, confers significant CO sensitivity to BK(Ca) channels. The rapid reversibility of CO and cyanide binding, coupled to information garnered from other CO-binding proteins, suggests that C911 may be involved in formation of a transition metal cluster which can bind and, thereafter, activate BK(Ca).


Subject(s)
Carbon Monoxide/metabolism , Cysteine/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Amino Acid Sequence , Calcium/metabolism , Cysteine/genetics , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Patch-Clamp Techniques , Potassium Cyanide/pharmacology
19.
J Cereb Blood Flow Metab ; 31(1): 102-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20424637

ABSTRACT

The exact etiology of delayed cerebral vasospasm following cerebral hemorrhage is not clear, but a family of compounds termed 'bilirubin oxidation end products (BOXes)' derived from heme has been implicated. As proper regulation of vascular smooth muscle tone involves large-conductance Ca(2+)- and voltage-dependent Slo1 K(+) (BK, maxiK, K(Ca)1.1) channels, we examined whether BOXes altered functional properties of the channel. Electrophysiological measurements of Slo1 channels heterologously expressed in a human cell line and of native mouse BK channels in isolated cerebral myocytes showed that BOXes markedly diminished open probability. Biophysically, BOXes specifically stabilized the conformations of the channel with its ion conduction gate closed. The results of chemical amino-acid modifications and molecular mutagenesis together suggest that two specific lysine residues in the structural element linking the transmembrane ion-permeation domain to the carboxyl cytosolic domain of the Slo1 channel are critical in determining the sensitivity of the channel to BOXes. Inhibition of Slo1 BK channels by BOXes may contribute to the development of delayed cerebral vasospasm following brain hemorrhage.


Subject(s)
Bilirubin/physiology , Cerebrovascular Circulation/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Muscle, Smooth, Vascular/physiology , Algorithms , Amino Acid Sequence , Animals , Basilar Artery/drug effects , Basilar Artery/metabolism , Bilirubin/cerebrospinal fluid , Bilirubin/metabolism , Electrophysiological Phenomena , Humans , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle Tonus/physiology , Oxidation-Reduction , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Vasospasm, Intracranial/physiopathology
20.
J Physiol ; 588(Pt 19): 3713-25, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20679351

ABSTRACT

Molecularly defined P2Y receptor subtypes are known to regulate the functions of neurons through an inhibition of K(V)7 K(+) and Ca(V)2 Ca(2+) channels and via an activation or inhibition of Kir3 channels. Here, we searched for additional neuronal ion channels as targets for P2Y receptors. Rat P2Y(1) receptors were expressed in PC12 cells via an inducible expression system, and the effects of nucleotides on membrane currents and intracellular Ca(2+) were investigated. At a membrane potential of 30 mV, ADP induced transient outward currents in a concentration-dependent manner with half-maximal effects at 4 µm. These currents had reversal potentials close to the K(+) equilibrium potential and changed direction when extracellular Na(+) was largely replaced by K(+), but remained unaltered when extracellular Cl() was changed. Currents were abolished by P2Y(1) antagonists and by blockade of phospholipase C. ADP also caused rises in intracellular Ca(2+), and ADP-evoked currents were abolished when inositol trisphosphate-sensitive Ca(2+) stores were depleted. Blockers of K(Ca)2, but not those of K(Ca)1.1 or K(Ca)3.1, channels largely reduced ADP-evoked currents. In hippocampal neurons, ADP also triggered outward currents at 30 mV which were attenuated by P2Y(1) antagonists, depletion of Ca(2+) stores, or a blocker of K(Ca)2 channels. These results demonstrate that activation of neuronal P2Y(1) receptors may gate Ca(2+)-dependent K(+) (K(Ca)2) channels via phospholipase C-dependent increases in intracellular Ca(2+) and thereby define an additional class of neuronal ion channels as novel effectors for P2Y receptors. This mechanism may form the basis for the control of synaptic plasticity via P2Y(1) receptors.


Subject(s)
Neurons/physiology , Potassium Channels, Calcium-Activated/physiology , Receptors, Purinergic P2Y1/physiology , Adenosine Diphosphate/pharmacology , Animals , Axons/physiology , Electrophysiological Phenomena , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Neurons/drug effects , PC12 Cells , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Receptors, Purinergic P2Y1/drug effects , Type C Phospholipases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...