Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
PLoS One ; 16(10): e0257896, 2021.
Article in English | MEDLINE | ID: mdl-34610026

ABSTRACT

INTRODUCTION: Peripheral artery disease (PAD) is a highly morbid condition in which impaired blood flow to the limbs leads to pain and tissue loss. Previously we identified 670 nm electromagnetic energy (R/NIR) to increase nitric oxide levels in cells and tissue. NO elicits relaxation of smooth muscle (SMC) by stimulating potassium efflux and membrane hyperpolarization. The actions of energy on ion channel activity have yet to be explored. Here we hypothesized R/NIR stimulates vasodilation through activation of potassium channels in SMC. METHODS: Femoral arteries or facial arteries from C57Bl/6 and Slo1-/- mice were isolated, pressurized to 60 mmHg, pre-constricted with U46619, and irradiated twice with energy R/NIR (10 mW/cm2 for 5 min) with a 10 min dark period between irradiations. Single-channel K+ currents were recorded at room temperature from cell-attached and excised inside-out membrane patches of freshly isolated mouse femoral arterial muscle cells using the patch-clamp technique. RESULTS: R/NIR stimulated vasodilation requires functional activation of the large conductance potassium channels. There is a voltage dependent outward current in SMC with light stimulation, which is due to increases in the open state probability of channel opening. R/NIR modulation of channel opening is eliminated pharmacologically (paxilline) and genetically (BKca α subunit knockout). There is no direct action of light to modulate channel activity as excised patches did not increase the open state probability of channel opening. CONCLUSION: R/NIR vasodilation requires indirect activation of the BKca channel.


Subject(s)
Electromagnetic Radiation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/radiation effects , Signal Transduction/radiation effects , Vasodilation/radiation effects , Animals , Electric Stimulation/methods , Electric Stimulation Therapy/methods , Femoral Artery/metabolism , Gene Knockout Techniques , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/radiation effects , Mice , Mice, Knockout , Nitric Oxide/metabolism , Patch-Clamp Techniques , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/therapy
2.
Cardiovasc Res ; 116(7): 1372-1385, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31738403

ABSTRACT

AIMS: The myogenic reactivity of the middle cerebral arteries (MCA) protects the brain by altering the diameter in response to changes in lumen pressure. Large conductance potassium (BK) channels are known to regulate the myogenic reactivity, yet, it is not clear how aging alters the myogenic reactivity via the BK channel in males and females. Thus, we hypothesize that age-associated changes in BK channel subunits modulate the myogenic reactivity in a sex-specific manner. METHODS AND RESULTS: We used vascular reactivity, patch-clamp, and biochemical methods to measure myogenic reactivity, BK channel function, and expression, respectively in cerebral vessels of adult and aged male and female Sprague Dawley rats. Our results suggest that aging and ovariectomy (OVX) exaggerated the myogenic reactivity of MCA in females but attenuated it in males. Aging induced outward eutrophic remodelling in females but inward hypertrophic remodelling in males. Aging decreased total, Kv, BK channel currents, and spontaneous transient outward currents (STOC) in vascular smooth muscle cells isolated from females, but not in males. Aging increased BKα subunit mRNA and protein both in males and females. However, aging decreased BKß1 subunit protein and mRNA in females only. In males, BKß1 mRNA is increased, but protein is decreased. Iberiotoxin-induced MCA constriction is lower in aged females but higher in aged males. Activation of BKα (10 µM NS1619) and BKß1 (10 µM S-Equol) subunits failed to increase STOCs and were unable to decrease the myogenic reactivity of MCA in aged female but not in aged male rats. OVX decreased, but chronic supplementation of oestradiol restored BK channel expression and function. CONCLUSION: Overall our results suggest that aging or OVX-associated downregulation of the BKß1 expression and function in females results in exaggerated myogenic reactivity of MCA. However, age-associated increase in BK channel function in males attenuated myogenic reactivity of MCA.


Subject(s)
Aging/metabolism , Cerebrovascular Circulation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Middle Cerebral Artery/metabolism , Vasoconstriction , Age Factors , Animals , Arterial Pressure , Female , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Membrane Potentials , Ovariectomy , Rats, Sprague-Dawley , Sex Factors , Signal Transduction , Vascular Remodeling
3.
J Med Food ; 22(9): 963-970, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31259654

ABSTRACT

Naringin and its aglycone, naringenin, occur naturally in our regular diet and traditional Chinese medicines. This study aimed to detect an effective therapeutic approach for cough variant asthma (CVA) through evaluating the relaxant effect of these two bioactive herbal monomers as antitussive and antiasthmatic on rat tracheal smooth muscle. The relaxant effect was determined by measuring muscular tension with a mechanical recording system in rat tracheal rings. Cytosolic Ca2+ concentration was measured using a confocal imaging system in primary cultured tracheal smooth muscle cells. In rat tracheal rings, addition of both naringin and naringenin could concentration dependently relax carbachol (CCh)-evoked tonic contraction. This epithelium-independent relaxation could be suppressed by BaCl2, tetraethylammonium, and iberiotoxin (IbTX), but not by glibenclamide. After stimulating primary cultured tracheal smooth muscle cells by CCh or high KCl, the intracellular Ca2+ increase could be inhibited by both naringin and naringenin, respectively. This reaction was also suppressed by IbTX. These results demonstrate that both naringin and naringenin can relax tracheal smooth muscle through opening big conductance Ca2+-activated K+ channel, which mediates plasma membrane hyperpolarization and reduces Ca2+ influx. Our data indicate a potentially effective therapeutic approach of naringin and naringenin for CVA.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Antitussive Agents/administration & dosage , Asthma/drug therapy , Flavanones/administration & dosage , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Plant Extracts/administration & dosage , Trachea/drug effects , Animals , Asthma/genetics , Asthma/metabolism , Asthma/physiopathology , Calcium/metabolism , Citrus/chemistry , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Male , Muscle Relaxation/drug effects , Rats , Rats, Sprague-Dawley , Trachea/physiopathology
4.
J Biol Chem ; 289(51): 35314-25, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25371198

ABSTRACT

Calcium/voltage-gated, large conductance potassium (BK) channels control numerous physiological processes, including myogenic tone. BK channel regulation by direct interaction between lipid and channel protein sites has received increasing attention. Leukotrienes (LTA4, LTB4, LTC4, LTD4, and LTE4) are inflammatory lipid mediators. We performed patch clamp studies in Xenopus oocytes that co-expressed BK channel-forming (cbv1) and accessory ß1 subunits cloned from rat cerebral artery myocytes. Leukotrienes were applied at 0.1 nm-10 µm to either leaflet of cell-free membranes at a wide range of [Ca(2+)]i and voltages. Only LTB4 reversibly increased BK steady-state activity (EC50 = 1 nm; Emax reached at 10 nm), with physiological [Ca(2+)]i and voltages favoring this activation. Homomeric cbv1 or cbv1-ß2 channels were LTB4-resistant. Computational modeling predicted that LTB4 docked onto the cholane steroid-sensing site in the BK ß1 transmembrane domain 2 (TM2). Co-application of LTB4 and cholane steroid did not further increase LTB4-induced activation. LTB4 failed to activate ß1 subunit-containing channels when ß1 carried T169A, A176S, or K179I within the docking site. Co-application of LTB4 with LTA4, LTC4, LTD4, or LTE4 suppressed LTB4-induced activation. Inactive leukotrienes docked onto a portion of the site, probably preventing tight docking of LTB4. In summary, we document the ability of two endogenous lipids from different chemical families to share their site of action on a channel accessory subunit. Thus, cross-talk between leukotrienes and cholane steroids might converge on regulation of smooth muscle contractility via BK ß1. Moreover, the identification of LTB4 as a highly potent ligand for BK channels is critical for the future development of ß1-specific BK channel activators.


Subject(s)
Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Leukotriene B4/metabolism , Animals , Calcium/metabolism , Cerebral Arteries/cytology , Female , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Leukotriene A4/chemistry , Leukotriene A4/metabolism , Leukotriene A4/pharmacology , Leukotriene B4/chemistry , Leukotriene B4/pharmacology , Leukotriene C4/chemistry , Leukotriene C4/metabolism , Leukotriene C4/pharmacology , Leukotriene D4/chemistry , Leukotriene D4/metabolism , Leukotriene D4/pharmacology , Leukotriene E4/chemistry , Leukotriene E4/metabolism , Leukotriene E4/pharmacology , Membrane Potentials/drug effects , Microinjections , Models, Molecular , Molecular Structure , Muscle Cells/cytology , Muscle Cells/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Protein Binding , Protein Structure, Tertiary , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Rats , Xenopus laevis
5.
Biochim Biophys Acta ; 1838(10): 2451-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24946143

ABSTRACT

Our previous study reported that cerebrosides from traditional Chinese medicine Baifuzi directly interact with the STREX domain of BKCa channels, which in turn results in the therapeutic effect of Baifuzi on ischemic stroke. However, it is not known how cerebrosides in the plasma membrane could interact with the STREX domain that is in the cytoplasmic side. Using patch-clamp technique, effects of different cerebrosides on the BKCa channel were studied by measuring single channel currents in CHO cells expressing wild type or mutated BKCa channels. Palmitoylation of the STREX domain was removed either by site-directed mutagenesis or pharmacological inhibition. Removal of palmitoylation sites at C646 and C647 by mutating the residues to Ala abolished the ability of cerebrosides to activate the BKCa channel. In contrast, the mutation neither changed the single channel conductance nor voltage sensitivity of the channel. Both palmitoylation inhibitors tunicamycin and palmitic acid analog 2-bromopalmitate attenuated the activation of the BKCa channel by cerebrosides. Furthermore, confocal images on STREX-EGFP fragments demonstrated that STREX fragments no longer associated with the plasma membrane when the palmitoylation was removed or blocked. These findings suggest that palmitoylation of the STREX domain is necessary for cerebrosides to activate the BKCa channel and provide insight into the mechanism of how Baifuzi could exert therapeutic effect on ischemic stroke.


Subject(s)
Avian Proteins/metabolism , Cell Membrane/metabolism , Cerebrosides/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Lipoylation/physiology , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , CHO Cells , Cell Membrane/chemistry , Cell Membrane/genetics , Cerebrosides/chemistry , Chickens , Cricetinae , Cricetulus , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Lipoylation/drug effects , Medicine, Chinese Traditional/methods , Mutagenesis, Site-Directed , Mutation, Missense , Patch-Clamp Techniques , Protein Structure, Tertiary , Stroke/drug therapy , Stroke/genetics , Stroke/metabolism
6.
PLoS One ; 8(6): e66278, 2013.
Article in English | MEDLINE | ID: mdl-23840433

ABSTRACT

BACKGROUND: Several data favor androgen receptor implication in prostate cancer initiation through the induction of several gene activation programs. The aim of the study is to identify potential biomarkers for early diagnosis of prostate cancer (PCa) among androgen-regulated genes (ARG) and to evaluate comparative expression of these genes in normal prostate and normal prostate-related androgen-sensitive tissues that do not (or rarely) give rise to cancer. METHODS: ARG were selected in non-neoplastic adult human prostatic epithelial RWPE-1 cells stably expressing an exogenous human androgen receptor, using RNA-microarrays and validation by qRT-PCR. Expression of 48 preselected genes was quantified in tissue samples (seminal vesicles, prostate transitional zones and prostate cancers, benign prostatic hypertrophy obtained from surgical specimens) using TaqMan® low-density arrays. The diagnostic performances of these potential biomarkers were compared to that of genes known to be associated with PCa (i.e. PCA3 and DLX1). RESULTS AND DISCUSSION: By crossing expression studies in 26 matched PCa and normal prostate transitional zone samples, and 35 matched seminal vesicle and PCa samples, 14 genes were identified. Similarly, 9 genes were overexpressed in 15 benign prostatic hypertrophy samples, as compared to PCa samples. Overall, we selected 8 genes of interest to evaluate their diagnostic performances in comparison with that of PCA3 and DLX1. Among them, 3 genes: CRYAB, KCNMA1 and SDPR, were overexpressed in all 3 reference non-cancerous tissues. The areas under ROC curves of these genes reached those of PCA3 (0.91) and DLX1 (0.94). CONCLUSIONS: We identified ARG with reduced expression in PCa and with significant diagnostic values for discriminating between cancerous and non-cancerous prostatic tissues, similar that of PCA3. Given their expression pattern, they could be considered as potentially protective against prostate cancer. Moreover, they could be complementary to known genes overexpressed in PCa and included along with them in multiplex diagnostic tools.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Prostatic Neoplasms/diagnosis , alpha-Crystallin B Chain/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Early Detection of Cancer , Gene Expression , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Male , Metribolone/pharmacology , Organ Specificity , Phosphate-Binding Proteins , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , ROC Curve , Real-Time Polymerase Chain Reaction , Seminal Vesicles/metabolism , Testosterone Congeners/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , alpha-Crystallin B Chain/genetics
7.
Proc Natl Acad Sci U S A ; 110(12): 4816-21, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487785

ABSTRACT

Long-chain polyunsaturated omega-3 fatty acids such as docosahexaenoic acid (DHA), found abundantly in oily fish, may have diverse health-promoting effects, potentially protecting the immune, nervous, and cardiovascular systems. However, the mechanisms underlying the purported health-promoting effects of DHA remain largely unclear, in part because molecular signaling pathways and effectors of DHA are only beginning to be revealed. In vascular smooth muscle cells, large-conductance Ca(2+)- and voltage-activated K(+) (BK) channels provide a critical vasodilatory influence. We report here that DHA with an EC50 of ∼500 nM rapidly and reversibly activates BK channels composed of the pore-forming Slo1 subunit and the auxiliary subunit ß1, increasing currents by up to ∼20-fold. The DHA action is observed in cell-free patches and does not require voltage-sensor activation or Ca(2+) binding but involves destabilization of the closed conformation of the ion conduction gate. DHA lowers blood pressure in anesthetized wild-type but not in Slo1 knockout mice. DHA ethyl ester, contained in dietary supplements, fails to activate BK channels and antagonizes the stimulatory effect of DHA. Slo1 BK channels are thus receptors for long-chain omega-3 fatty acids, and these fatty acids--unlike their ethyl ester derivatives--activate the channels and lower blood pressure. This finding has practical implications for the use of omega-3 fatty acids as nutraceuticals for the general public and also for the critically ill receiving omega-3-enriched formulas.


Subject(s)
Blood Pressure/drug effects , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Vasodilation/drug effects , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/drug effects , Mice , Mice, Knockout , Protein Structure, Tertiary
8.
Proc Natl Acad Sci U S A ; 108(26): 10744-9, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670298

ABSTRACT

Protein myristoylation is a means by which cells anchor proteins into membranes. The most common type of myristoylation occurs at an N-terminal glycine. However, myristoylation rarely occurs at an internal amino acid residue. Here we tested whether the α-subunit of the human large-conductance voltage- and Ca(2+)-activated K(+) channel (hSlo1) might undergo internal myristoylation. hSlo1 expressed in HEK293T cells incorporated [(3)H]myristic acid via a posttranslational mechanism, which is insensitive to cycloheximide, an inhibitor of protein biosynthesis. In-gel hydrolysis of [(3)H]myristoyl-hSlo1 with alkaline NH(2)OH (which cleaves hydroxyesters) but not neutral NH(2)OH (which cleaves thioesters) completely removed [(3)H]myristate from hSlo1, suggesting the involvement of a hydroxyester bond between hSlo1's hydroxyl-bearing serine, threonine, and/or tyrosine residues and myristic acid; this type of esterification was further confirmed by its resistance to alkaline Tris·HCl. Treatment of cells expressing hSlo1 with 100 µM myristic acid caused alteration of hSlo1 activation kinetics and a 40% decrease in hSlo1 current density from 20 to 12 nA*MΩ. Immunocytochemistry confirmed a decrease in hSlo1 plasmalemma localization by myristic acid. Replacement of the six serines or the seven threonines (but not of the single tyrosine) of hSlo1 intracellular loops 1 and 3 with alanines decreased hSlo1 direct myristoylation by 40-44%, whereas in combination decreased myristoylation by nearly 90% and abolished the myristic acid-induced change in current density. Our data demonstrate that an ion channel, hSlo1, is internally and posttranslationally myristoylated. Myristoylation occurs mainly at hSlo1 intracellular loop 1 or 3, and is an additional mechanism for channel surface expression regulation.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myristic Acid/metabolism , Serine/metabolism , Threonine/metabolism , Cell Line , Cell Membrane/metabolism , Esters , Humans , Immunohistochemistry , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Mutation , Patch-Clamp Techniques , Protein Processing, Post-Translational , Threonine/chemistry
9.
Shock ; 35(5): 485-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21330953

ABSTRACT

Nitric oxide-mediated activation of large conductance calcium-activated potassium (BK) channels is considered an important underlying mechanism of sepsis-induced hypotension. Indeed, the nonselective K-channel inhibitor, tetraethylammonium chloride (TEA), has been proposed as a potential treatment to raise blood pressure in septic shock by virtue of its ability to inhibit BK channels. As experimental evidence has so far relied on pharmacological inhibition, we examined the effects of channel deletion using BKα subunit knockout (α, Slo) mice in two mouse models of polymicrobial sepsis, namely, intraperitoneal fecal slurry and cecal ligation and puncture. Comparison was made against TEA treatment in wild-type (WT) mice. Following slurry, BKα and WT mice developed similar degrees of hypotension over 10 h with no difference in cardiac output as assessed by echocardiography between groups. Tetraethylammonium chloride raised blood pressure significantly in septic WT mice, but had no effect on survival. However, following cecal ligation and puncture, a significantly reduced survival was seen in both BKα mice and (high-dose) TEA-treated WT mice compared with untreated WT animals. In conclusion, the BK channel does not appear to be integral to sepsis-induced hypotension but does affect survival through other mechanisms. The pressor effect of TEA may be related to effects on other potassium channels.


Subject(s)
Hypotension/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/deficiency , Sepsis/microbiology , Sepsis/mortality , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Genotype , Hypotension/drug therapy , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Male , Mice , Mice, Knockout , Potassium Channel Blockers/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/drug therapy , Sepsis/genetics , Tetraethylammonium/therapeutic use
10.
Proc Natl Acad Sci U S A ; 107(44): 19096-101, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20959415

ABSTRACT

Large conductance voltage- and calcium-activated potassium channels (MaxiK, BK(Ca)) are well known for sustaining cerebral and coronary arterial tone and for their linkage to vasodilator ß-adrenergic receptors. However, how MaxiK channels are linked to counterbalancing vasoconstrictor receptors is unknown. Here, we show that vasopressive thromboxane A2 receptors (TP) can intimately couple with and inhibit MaxiK channels. Activation of the receptor with its agonist trans-inhibits MaxiK independently of G-protein activation. This unconventional mechanism is supported by independent lines of evidence: (i) inhibition of MaxiK current by thromboxane A2 mimetic, U46619, occurs even when G-protein activity is suppressed; (ii) MaxiK and TP physically associate and display a high degree of proximity; and (iii) Förster resonance energy transfer occurs between fluorescently labeled MaxiK and TP, supporting a direct interaction. The molecular mechanism of MaxiK-TP intimate interaction involves the receptor's first intracellular loop and C terminus, and it entails the voltage-sensing conduction cassette of MaxiK channel. Further, physiological evidence of MaxiK-TP physical interaction is given in human coronaries and rat aorta, and by confirming TP role (with antagonist SQ29,548) in the U46619-induced MaxiK inhibition in human coronaries. We propose that vasoconstrictor TP receptor and MaxiK-channel direct interaction facilitates G-protein-independent TP to MaxiK trans-inhibition, which would promote vasoconstriction.


Subject(s)
Aorta/metabolism , Coronary Vessels/metabolism , GTP-Binding Proteins/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic , Enzyme Activation , Fatty Acids, Unsaturated , GTP-Binding Proteins/genetics , Humans , Hydrazines/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, Thromboxane A2, Prostaglandin H2/agonists , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
11.
J Physiol ; 588(Pt 10): 1763-77, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20351045

ABSTRACT

Colonic epithelial K(+) secretion is a two-step transport process with initial K(+) uptake over the basolateral membrane followed by K(+) channel-dependent exit into the lumen. In this process the large-conductance, Ca(2+)-activated K(Ca)1.1 (BK) channel has been identified as the only apparent secretory K(+) channel in the apical membrane of the murine distal colon. The BK channel is responsible for both resting and Ca(2+)-activated colonic K(+) secretion and is up-regulated by aldosterone. Agonists (e.g. adrenaline) that elevate cAMP are potent activators of distal colonic K(+) secretion. However, the secretory K(+) channel responsible for cAMP-induced K(+) secretion remains to be defined. In this study we used the Ussing chamber to identify adrenaline-induced electrogenic K(+) secretion. We found that the adrenaline-induced electrogenic ion secretion is a compound effect dominated by anion secretion and a smaller electrically opposing K(+) secretion. Using tissue from (i) BK wildtype (BK(+/+)) and knockout (BK(/)) and (ii) cystic fibrosis transmembrane regulator (CFTR) wildtype (CFTR(+/+)) and knockout (CFTR(/)) mice we were able to isolate the adrenaline-induced K(+) secretion. We found that adrenaline-induced K(+) secretion: (1) is absent in colonic epithelia from BK(/) mice, (2) is greatly up-regulated in mice on a high K(+) diet and (3) is present as sustained positive current in colonic epithelia from CFTR(/) mice. We identified two known C-terminal BK alpha-subunit splice variants in colonic enterocytes (STREX and ZERO). Importantly, the ZERO variant known to be activated by cAMP is differentially up-regulated in enterocytes from animals on a high K(+) diet. In summary, these results strongly suggest that the adrenaline-induced distal colonic K(+) secretion is mediated by the BK channel and probably involves aldosterone-induced ZERO splice variant up-regulation.


Subject(s)
Colon/metabolism , Epinephrine/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Potassium Channel Blockers , Potassium/metabolism , Adrenergic beta-Antagonists/pharmacology , Aldosterone/blood , Animals , Colon/drug effects , Cyclic AMP/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/isolation & purification , Diffusion Chambers, Culture , Electrophysiology , Enterocytes/drug effects , Enterocytes/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Mice , Mice, Knockout , Potassium/pharmacology , Potassium, Dietary/pharmacology , Propranolol/pharmacology , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
12.
J Gen Physiol ; 132(1): 13-28, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18562499

ABSTRACT

Large conductance, calcium- and voltage-gated potassium (BK) channels are ubiquitous and critical for neuronal function, immunity, and smooth muscle contractility. BK channels are thought to be regulated by phosphatidylinositol 4,5-bisphosphate (PIP(2)) only through phospholipase C (PLC)-generated PIP(2) metabolites that target Ca(2+) stores and protein kinase C and, eventually, the BK channel. Here, we report that PIP(2) activates BK channels independently of PIP(2) metabolites. PIP(2) enhances Ca(2+)-driven gating and alters both open and closed channel distributions without affecting voltage gating and unitary conductance. Recovery from activation was strongly dependent on PIP(2) acyl chain length, with channels exposed to water-soluble diC4 and diC8 showing much faster recovery than those exposed to PIP(2) (diC16). The PIP(2)-channel interaction requires negative charge and the inositol moiety in the phospholipid headgroup, and the sequence RKK in the S6-S7 cytosolic linker of the BK channel-forming (cbv1) subunit. PIP(2)-induced activation is drastically potentiated by accessory beta(1) (but not beta(4)) channel subunits. Moreover, PIP(2) robustly activates BK channels in vascular myocytes, where beta(1) subunits are abundantly expressed, but not in skeletal myocytes, where these subunits are barely detectable. These data demonstrate that the final PIP(2) effect is determined by channel accessory subunits, and such mechanism is subunit specific. In HEK293 cells, cotransfection of cbv1+beta(1) and PI4-kinaseIIalpha robustly activates BK channels, suggesting a role for endogenous PIP(2) in modulating channel activity. Indeed, in membrane patches excised from vascular myocytes, BK channel activity runs down and Mg-ATP recovers it, this recovery being abolished by PIP(2) antibodies applied to the cytosolic membrane surface. Moreover, in intact arterial myocytes under physiological conditions, PLC inhibition on top of blockade of downstream signaling leads to drastic BK channel activation. Finally, pharmacological treatment that raises PIP(2) levels and activates BK channels dilates de-endothelized arteries that regulate cerebral blood flow. These data indicate that endogenous PIP(2) directly activates vascular myocyte BK channels to control vascular tone.


Subject(s)
Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Signal Transduction/physiology , Adenosine Triphosphate/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Calcium/pharmacology , Electrophysiology , Enzyme Inhibitors/pharmacology , Female , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Middle Cerebral Artery/cytology , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/physiology , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Okadaic Acid/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphatidylinositol Phosphates/pharmacology , Phosphatidylinositols/pharmacology , Polylysine/pharmacology , RNA, Complementary/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Xenopus laevis
13.
J Gen Physiol ; 131(2): 147-61, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18227273

ABSTRACT

The internal vestibule of large-conductance Ca(2+) voltage-activated K(+) (BK) channels contains a ring of eight negative charges not present in K(+) channels of lower conductance (Glu386 and Glu389 in hSlo) that modulates channel conductance through an electrostatic mechanism (Brelidze, T.I., X. Niu, and K.L. Magleby. 2003. Proc. Natl. Acad. Sci. USA. 100:9017-9022). In BK channels there are also two acidic amino acid residues in an extracellular loop (Asp326 and Glu329 in hSlo). To determine the electrostatic influence of these charges on channel conductance, we expressed wild-type BK channels and mutants E386N/E389N, D326N, E329Q, and D326N/E329Q channels on Xenopus laevis oocytes, and measured the expressed currents under patch clamp. Contribution of E329 to the conductance is negligible and single channel conductance of D326N/E329Q channels measured at 0 mV in symmetrical 110 mM K(+) was 18% lower than the control. Current-voltage curves displayed weak outward rectification for D326N and the double mutant. The conductance differences between the mutants and wild-type BK were caused by an electrostatic effect since they were enhanced at low K(+) (30 mM) and vanished at high K(+) (1 M K(+)). We determine the electrostatic potential change, Deltaphi, caused by the charge neutralization using TEA(+) block for the extracellular charges and Ba(2+) for intracellular charges. We measured 13 +/- 2 mV for Deltaphi at the TEA(+) site when turning off the extracellular charges, and 17 +/- 2 mV for the Deltaphi at the Ba(2+) site when the intracellular charges were turned off. To understand the electrostatic effect of charge neutralizations, we determined Deltaphi using a BK channel molecular model embedded in a lipid bilayer and solving the Poisson-Boltzmann equation. The model explains the experimental results adequately and, in particular, gives an economical explanation to the differential effect on the conductance of the neutralization of charges D326 and E329.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Mutation , Algorithms , Amino Acid Sequence , Amino Acid Substitution , Animals , Barium/pharmacology , Calcium Channel Blockers/pharmacology , Charybdotoxin/pharmacology , Computer Simulation , Female , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/genetics , Membrane Potentials/physiology , Models, Molecular , Molecular Sequence Data , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Osmolar Concentration , Patch-Clamp Techniques , Potassium/metabolism , Potassium/pharmacology , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Sequence Homology, Amino Acid , Static Electricity , Tetraethylammonium/pharmacology , Xenopus laevis
14.
J Gen Physiol ; 128(4): 389-404, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17001085

ABSTRACT

The activation of BK channels by Ca(2+) is highly cooperative, with small changes in intracellular Ca(2+) concentration having large effects on open probability (Po). Here we examine the mechanism of cooperative activation of BK channels by Ca(2+). Each of the four subunits of BK channels has a large intracellular COOH terminus with two different high-affinity Ca(2+) sensors: an RCK1 sensor (D362/D367) located on the RCK1 (regulator of conductance of K(+)) domain and a Ca-bowl sensor located on or after the RCK2 domain. To determine interactions among these Ca(2+) sensors, we examine channels with eight different configurations of functional high-affinity Ca(2+) sensors on the four subunits. We find that the RCK1 sensor and Ca bowl contribute about equally to Ca(2+) activation of the channel when there is only one high-affinity Ca(2+) sensor per subunit. We also find that an RCK1 sensor and a Ca bowl on the same subunit are much more effective in increasing Po than when they are on different subunits, indicating positive intrasubunit cooperativity. If it is assumed that BK channels have a gating ring similar to MthK channels with alternating RCK1 and RCK2 domains and that the Ca(2+) sensors act at the flexible (rather than fixed) interfaces between RCK domains, then a comparison of the distribution of Ca(2+) sensors with the observed responses suggest that the interface between RCK1 and RCK2 domains on the same subunit is flexible. On this basis, intrasubunit cooperativity arises because two high-affinity Ca(2+) sensors acting across a flexible interface are more effective in opening the channel than when acting at separate interfaces. An allosteric model incorporating intrasubunit cooperativity nested within intersubunit cooperativity could approximate the Po vs. Ca(2+) response for eight possible subunit configurations of the high-affinity Ca(2+) sensors as well as for three additional configurations from a previous study.


Subject(s)
Calcium/metabolism , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Algorithms , Allosteric Regulation , Animals , Binding Sites/genetics , Female , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Models, Biological , Mutation/genetics , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/physiology , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Xenopus laevis
15.
J Gen Physiol ; 128(4): 423-41, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16966472

ABSTRACT

Crystal structures of potassium channels have strongly corroborated an earlier hypothetical picture based on functional studies, in which the channel gate was located on the cytoplasmic side of the pore. However, accessibility studies on several types of ligand-sensitive K(+) channels have suggested that their activation gates may be located near or within the selectivity filter instead. It remains to be determined to what extent the physical location of the gate is conserved across the large K(+) channel family. Direct evidence about the location of the gate in large conductance calcium-activated K(+) (BK) channels, which are gated by both voltage and ligand (calcium), has been scarce. Our earlier kinetic measurements of the block of BK channels by internal quaternary ammonium ions have raised the possibility that they may lack a cytoplasmic gate. We show in this study that a synthesized Shaker ball peptide (ShBP) homologue acts as a state-dependent blocker for BK channels when applied internally, suggesting a widening at the intracellular end of the channel pore upon gating. This is consistent with a gating-related conformational change at the cytoplasmic end of the pore-lining helices, as suggested by previous functional and structural studies on other K(+) channels. Furthermore, our results from two BK channel mutations demonstrate that similar types of interactions between ball peptides and channels are shared by BK and other K(+) channel types.


Subject(s)
Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Animals , Calcium/pharmacology , Female , Intracellular Signaling Peptides and Proteins , Ion Channel Gating/drug effects , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mutation/genetics , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Quaternary Ammonium Compounds/pharmacology , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/physiology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL