Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Commun Biol ; 4(1): 1392, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907346

ABSTRACT

Plasma membrane phosphatidylinositol 4-phosphate (PI4P) is a precursor of PI(4,5)P2, an important regulator of a large number of ion channels. Although the role of the phospholipid PI(4,5)P2 in stabilizing ion channel function is well established, little is known about the role of phospholipids in channel membrane localization and specifically the role of PI4P in channel function and localization. The phosphatidylinositol 4-kinases (PI4Ks) synthesize PI4P. Our data show that inhibition of PI4K and prolonged decrease of levels of plasma membrane PI4P lead to a decrease in the KCNQ1/KCNE1 channel membrane localization and function. In addition, we show that mutations linked to Long QT syndrome that affect channel interactions with phospholipids lead to a decrease in membrane expression. We show that expression of a LQT1-associated C-terminal deletion mutant abolishes PI4Kinase-mediated decrease in membrane expression and rescues membrane expression for phospholipid-targeting mutations. Our results indicate a novel role for PI4P on ion channel regulation. Our data suggest that decreased membrane PI4P availability to the channel, either due to inhibition of PI4K or as consequence of mutations, dramatically inhibits KCNQ1/KCNE1 channel membrane localization and current. Our results may have implications to regulation of other PI4P binding channels.


Subject(s)
Cell Membrane/metabolism , KCNQ1 Potassium Channel/genetics , Phosphatidylinositol Phosphates/metabolism , Potassium Channels, Voltage-Gated/genetics , Animals , Female , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Rats , Rats, Sprague-Dawley
2.
PLoS One ; 15(8): e0237591, 2020.
Article in English | MEDLINE | ID: mdl-32833978

ABSTRACT

The slow cardiac delayed rectifier current (IKs) is formed by KCNQ1 and KCNE1 subunits and is one of the major repolarizing currents in the heart. Decrease of IKs currents either due to inherited mutations or pathological remodeling is associated with increased risk for cardiac arrhythmias and sudden death. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in heart disease and diabetes. Recently, we found that sustained stimulation of the calcium-dependent PKCßII isoform leads to decrease in KCNQ1 subunit membrane localization and KCNQ1/KCNE1 channel activity, although the role of KCNE1 in this regulation was not explored. Here, we show that the auxiliary KCNE1 subunit expression is necessary for channel internalization. A mutation in a KCNE1 phosphorylation site (KCNE1(S102A)) abolished channel internalization in both heterologous expression systems and cardiomyocytes. Altogether, our results suggest that KCNE1(S102) phosphorylation by PKCßII leads to KCNQ1/KCNE1 channel internalization in response to sustained PKC stimulus, while leaving KCNQ1 homomeric channels in the membrane. This preferential internalization is expected to have strong impact on cardiac repolarization. Our results suggest that KCNE1(S102) is an important anti-arrhythmic drug target to prevent IKs pathological remodeling leading to cardiac arrhythmias.


Subject(s)
Calcium/metabolism , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Kinase C/metabolism , Animals , Female , HEK293 Cells , Humans , KCNQ1 Potassium Channel/genetics , Mutation , Myocytes, Cardiac/cytology , Potassium Channels, Voltage-Gated/genetics , Protein Kinase C/genetics , Rats
3.
Biochem Biophys Res Commun ; 465(3): 464-70, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26277396

ABSTRACT

Protein kinase C (PKC) plays key roles in the regulation of signal transduction and cellular function in various cell types. At least ten PKC isoforms have been identified and intracellular localization and trafficking of these individual isoforms are important for regulation of enzyme activity and substrate specificity. PKC can be activated downstream of Gq-protein coupled receptor (GqPCR) signaling and translocate to various cellular compartments including plasma membrane (PM). Recent reports suggested that different types of GqPCRs would activate different PKC isoforms (classic, novel and atypical PKCs) with different trafficking patterns. However, the knowledge of isoform-specific activation of PKC by each GqPCR is limited. α1-Adrenoceptor (α1-AR) is one of the GqPCRs highly expressed in the cardiovascular system. In this study, we examined the isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-AR stimulation (α1-ARS). Rat PKCα, ßI, ßII, δ, ε and ζ fused with GFP at C-term were co-transfected with human α1A-AR into HEK293T cells. The isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-ARS using phenylephrine was measured by confocal microscopy. Before stimulation, GFP-PKCs were localized at cytosolic region. α1-ARS strongly and rapidly translocated a classical PKC (cPKC), PKCα, (<30 s) to PM, with PKCα returning diffusively into the cytosol within 5 min. α1-ARS rapidly translocated other cPKCs, PKCßI and PKCßII, to the PM (<30 s), with sustained membrane localization. One novel PKC (nPKC), PKCε, but not another nPKC, PKCδ, was translocated by α1-AR stimulation to the PM (<30 s) and its membrane localization was also sustained. Finally, α1-AR stimulation did not cause a diacylglycerol-insensitive atypical PKC, PKCζ translocation. Our data suggest that PKCα, ß and ε activation may underlie physiological and pathophysiological responses of α1-AR signaling for the phosphorylation of membrane-associated substrates including ion-channel and transporter proteins in the cardiovascular system.


Subject(s)
Cell Membrane/metabolism , Protein Kinase C/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/physiology , HEK293 Cells , Humans , Protein Isoforms/metabolism , Protein Transport/physiology
4.
Circ Res ; 110(1): 59-70, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22076634

ABSTRACT

RATIONALE: The Rad-Gem/Kir-related family (RGKs) consists of small GTP-binding proteins that strongly inhibit the activity of voltage-gated calcium channels. Among RGKs, Rem1 is strongly and specifically expressed in cardiac tissue. However, the physiological role and regulation of RGKs, and Rem1 in particular, are largely unknown. OBJECTIVE: To determine if Rem1 function is physiologically regulated by adrenergic signaling and thus impacts voltage-gated L-type calcium channel (VLCC) activity in the heart. METHODS AND RESULTS: We found that activation of protein kinase D1, a protein kinase downstream of α(1)-adrenergic signaling, leads to direct phosphorylation of Rem1 at Ser18. This results in an increase of the channel activity and plasma membrane expression observed by using a combination of electrophysiology, live cell confocal microscopy, and immunohistochemistry in heterologous expression system and neonatal cardiomyocytes. In addition, we show that stimulation of α(1)-adrenergic receptor-protein kinase D1-Rem1 signaling increases transverse-tubule VLCC expression that results in increased L-type Ca(2+) current density in adult ventricular myocytes. CONCLUSION: The α(1)-adrenergic stimulation releases Rem1 inhibition of VLCCs through direct phosphorylation of Rem1 at Ser18 by protein kinase D1, resulting in an increase of the channel activity and transverse-tubule expression. Our results uncover a novel molecular regulatory mechanism of VLCC trafficking and function in the heart and provide the first demonstration of physiological regulation of RGK function.


Subject(s)
Calcium Channels, L-Type/physiology , Myocytes, Cardiac/physiology , Protein Kinases/physiology , Protein Transport/physiology , Receptors, Adrenergic, alpha-1/physiology , Signal Transduction/physiology , Animals , Cell Membrane/physiology , Cells, Cultured , Male , Microtubules/physiology , Models, Animal , Monomeric GTP-Binding Proteins/physiology , Myocytes, Cardiac/cytology , Patch-Clamp Techniques , Phosphorylation , Protein Kinase C , Rats , Rats, Sprague-Dawley
5.
Sci Transl Med ; 3(76): 76ra28, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21451124

ABSTRACT

Inherited long QT syndrome (LQTS) is caused by mutations in ion channels that delay cardiac repolarization, increasing the risk of sudden death from ventricular arrhythmias. Currently, the risk of sudden death in individuals with LQTS is estimated from clinical parameters such as age, gender, and the QT interval, measured from the electrocardiogram. Even though a number of different mutations can cause LQTS, mutation-specific information is rarely used clinically. LQTS type 1 (LQT1), one of the most common forms of LQTS, is caused by mutations in the slow potassium current (I(Ks)) channel α subunit KCNQ1. We investigated whether mutation-specific changes in I(Ks) function can predict cardiac risk in LQT1. By correlating the clinical phenotype of 387 LQT1 patients with the cellular electrophysiological characteristics caused by an array of mutations in KCNQ1, we found that channels with a decreased rate of current activation are associated with increased risk of cardiac events (hazard ratio=2.02), independent of the clinical parameters usually used for risk stratification. In patients with moderate QT prolongation (a QT interval less than 500 ms), slower activation was an independent predictor for cardiac events (syncope, aborted cardiac arrest, and sudden death) (hazard ratio = 2.10), whereas the length of the QT interval itself was not. Our results indicate that genotype and biophysical phenotype analysis may be useful for risk stratification of LQT1 patients and suggest that slow channel activation is associated with an increased risk of cardiac events.


Subject(s)
Ion Channel Gating/physiology , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Mutation , Adolescent , Adult , Animals , Child , Child, Preschool , Computer Simulation , Electrophysiology , Genetic Predisposition to Disease , Genotype , Humans , Infant , Kaplan-Meier Estimate , Male , Models, Biological , Oocytes/cytology , Oocytes/physiology , Phenotype , Proportional Hazards Models , Registries , Risk Factors , Xenopus laevis , Young Adult
6.
J Cardiovasc Electrophysiol ; 22(2): 193-200, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20662986

ABSTRACT

UNLABELLED: BACKGROUND: Data regarding possible ion channel mechanisms that predispose to ventricular tachyarrhythmias in patients with phenotype-negative long-QT syndrome (LQTS) are limited. METHODS AND RESULTS: We carried out cellular expression studies for the S349W mutation in the KCNQ1 channel, which was identified in 15 patients from the International LQTS Registry who experienced a high rate of cardiac events despite lack of significant QTc prolongation. The clinical outcome of S349W mutation carriers was compared with that of QTc-matched carriers of haploinsufficient missense (n = 30) and nonsense (n = 45) KCNQ1 mutations. The channels containing the mutant S349W subunit showed a mild reduction in current (<50%), in the haploinsuficient range, with an increase in maximal conductance compared with wild-type channels. In contrast, expression of the S349W mutant subunit produced a pronounced effect on both the voltage dependence of activation and the time constant of activation, while haploinsuficient channels showed no effect on either parameter. The cumulative probability of cardiac events from birth through age 20 years was significantly higher among S349W mutation carriers (58%) as compared with carriers of QTc-matched haploinsufficent missense (21%, P = 0.004) and nonsense (25%, P = 0.01) mutations. CONCLUSIONS: The S349W mutation in the KCNQ1 potassium channel exerts a relatively mild effect on the ion channel current, whereas an increase in conductance compensates for impaired voltage activation of the channel. The changes observed in voltage activation of the channel may underlie the mechanisms predisposing to arrhythmic risk among LQTS patients with a normal-range QTc.


Subject(s)
Death, Sudden, Cardiac , Genetic Predisposition to Disease/genetics , Ion Channel Gating/genetics , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/genetics , Child , Female , Genotype , Humans , Male , Phenotype , Polymorphism, Single Nucleotide
7.
J Am Soc Nephrol ; 21(12): 2117-29, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21088294

ABSTRACT

SeSAME/EAST syndrome is a channelopathy consisting of a hypokalemic, hypomagnesemic, metabolic alkalosis associated with seizures, sensorineural deafness, ataxia, and developmental abnormalities. This disease links to autosomal recessive mutations in KCNJ10, which encodes the Kir4.1 potassium channel, but the functional consequences of these mutations are not well understood. In Xenopus oocytes, all of the disease-associated mutant channels (R65P, R65P/R199X, G77R, C140R, T164I, and A167V/R297C) had decreased K(+) current (0 to 23% of wild-type levels). Immunofluorescence demonstrated decreased surface expression of G77R, C140R, and A167V expressed in HEK293 cells. When we coexpressed mutant and wild-type subunits to mimic the heterozygous state, R199X, C140R, and G77R currents decreased to 55, 40, and 20% of wild-type levels, respectively, suggesting that carriers of these mutations may present with an abnormal phenotype. Because Kir4.1 subunits can form heteromeric channels with Kir5.1, we coexpressed the aforementioned mutants with Kir5.1 and found that currents were reduced at least as much as observed when we expressed mutants alone. Reduction of pH(i) from approximately 7.4 to 6.8 significantly decreased currents of all mutants except R199X but did not affect wild-type channels. In conclusion, perturbed pH gating may underlie the loss of channel function for the disease-associated mutant Kir4.1 channels and may have important physiologic consequences.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hearing Loss, Sensorineural/genetics , Kv1.1 Potassium Channel/genetics , Mutation , Seizures/genetics , Alkalosis/genetics , Alkalosis/physiopathology , Analysis of Variance , Animals , Ataxia/genetics , Ataxia/physiopathology , Fluorescent Antibody Technique , Genetic Predisposition to Disease , HEK293 Cells/metabolism , Hearing Loss, Sensorineural/physiopathology , Humans , Hypokalemia/genetics , Hypokalemia/physiopathology , Immunoblotting , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Models, Animal , Molecular Biology , Oocytes , Seizures/physiopathology , Syndrome , Xenopus laevis
9.
Circ Res ; 107(4): 532-9, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20576935

ABSTRACT

RATIONALE: Excess signaling through cardiac Gbetagamma subunits is an important component of heart failure (HF) pathophysiology. They recruit elevated levels of cytosolic G protein-coupled receptor kinase (GRK)2 to agonist-stimulated beta-adrenergic receptors (beta-ARs) in HF, leading to chronic beta-AR desensitization and downregulation; these events are all hallmarks of HF. Previous data suggested that inhibiting Gbetagamma signaling and its interaction with GRK2 could be of therapeutic value in HF. OBJECTIVE: We sought to investigate small molecule Gbetagamma inhibition in HF. METHODS AND RESULTS: We recently described novel small molecule Gbetagamma inhibitors that selectively block Gbetagamma-binding interactions, including M119 and its highly related analog, gallein. These compounds blocked interaction of Gbetagamma and GRK2 in vitro and in HL60 cells. Here, we show they reduced beta-AR-mediated membrane recruitment of GRK2 in isolated adult mouse cardiomyocytes. Furthermore, M119 enhanced both adenylyl cyclase activity and cardiomyocyte contractility in response to beta-AR agonist. To evaluate their cardiac-specific effects in vivo, we initially used an acute pharmacological HF model (30 mg/kg per day isoproterenol, 7 days). Concurrent daily injections prevented HF and partially normalized cardiac morphology and GRK2 expression in this acute HF model. To investigate possible efficacy in halting progression of preexisting HF, calsequestrin cardiac transgenic mice (CSQ) with extant HF received daily injections for 28 days. The compound alone halted HF progression and partially normalized heart size, morphology, and cardiac expression of HF marker genes (GRK2, atrial natriuretic factor, and beta-myosin heavy chain). CONCLUSIONS: These data suggest a promising therapeutic role for small molecule inhibition of pathological Gbetagamma signaling in the treatment of HF.


Subject(s)
GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Heart Failure/prevention & control , Signal Transduction/physiology , Animals , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , Disease Progression , Female , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , HL-60 Cells , Heart Failure/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac , Signal Transduction/drug effects , Xanthenes/pharmacology , Xanthenes/therapeutic use , Xenopus
10.
Circ Res ; 106(7): 1190-6, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20185796

ABSTRACT

RATIONALE: The mitochondrial ATP sensitive potassium channel (mK(ATP)) is implicated in cardioprotection by ischemic preconditioning (IPC), but the molecular identity of the channel remains controversial. The validity of current methods to assay mK(ATP) activity is disputed. OBJECTIVE: We sought to develop novel methods to assay mK(ATP) activity and its regulation. METHODS AND RESULTS: Using a thallium (Tl(+))-sensitive fluorophore, we developed a novel Tl(+) flux based assay for mK(ATP) activity, and used this assay probe several aspects of mK(ATP) function. The following key observations were made. (1) Time-dependent run down of mK(ATP) activity was reversed by phosphatidylinositol-4,5-bisphosphate (PIP(2)). (2) Dose responses of mK(ATP) to nucleotides revealed a UDP EC(50) of approximately 20 micromol/L and an ATP IC(50) of approximately 5 micromol/L. (3) The antidepressant fluoxetine (Prozac) inhibited mK(ATP) (IC(50)=2.4 micromol/L). Fluoxetine also blocked cardioprotection triggered by IPC, but did not block protection triggered by a mK(ATP)-independent stimulus. The related antidepressant zimelidine was without effect on either mK(ATP) or IPC. CONCLUSIONS: The Tl(+) flux mK(ATP) assay was validated by correlation with a classical mK(ATP) channel osmotic swelling assay (R(2)=0.855). The pharmacological profile of mK(ATP) (response to ATP, UDP, PIP(2), and fluoxetine) is consistent with that of an inward rectifying K(+) channel (K(IR)) and is somewhat closer to that of the K(IR)6.2 than the K(IR)6.1 isoform. The effect of fluoxetine on mK(ATP)-dependent cardioprotection has implications for the growing use of antidepressants in patients who may benefit from preconditioning.


Subject(s)
Biological Assay/methods , Mitochondria, Heart/metabolism , Potassium Channels/metabolism , Potassium/metabolism , Spectrometry, Fluorescence , Thallium/metabolism , Adenosine Triphosphate/metabolism , Animals , Antidepressive Agents, Second-Generation/pharmacology , Benzothiazoles , Coumarins , Fluorescent Dyes , Fluoxetine/pharmacology , Glycine/analogs & derivatives , In Vitro Techniques , Ischemic Preconditioning, Myocardial , Kinetics , Male , Mitochondria, Heart/drug effects , Mitochondrial Swelling , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Osmosis , Perfusion , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels/drug effects , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Uridine Diphosphate/metabolism
11.
Channels (Austin) ; 4(1): 3-11, 2010.
Article in English | MEDLINE | ID: mdl-19934648

ABSTRACT

Long-QT syndrome causes torsade de pointes arrhythmia, ventricular fibrillation, and sudden death. The most commonly inherited form of long-QT syndrome, LQT1, is due to mutations on the potassium channel gene KCNQ1, which forms one of the main repolarizing cardiac K(+) channels, IKs. IKs has been shown to be regulated by both beta-adrenergic receptors, via protein kinase A (PKA), and by Gq protein coupled receptors (GqPCR), via protein kinase C (PKC) and phosphatidylinositol 4,5-bisphosphate (PIP(2)). These regulatory pathways were shown to crosstalk, with PKA phosphorylation increasing the apparent affinity of IKs to PIP(2). Here we study the effects of LQT1 mutations in putative PIP(2)-KCNQ1 interaction sites on regulation of IKs by PKA and GqPCR. The effect of the LQT1 mutations on IKs regulation was tested for mutations in conserved, positively charged amino acids, located in four distinct cytoplamic domains of the KCNQ1 subunit: R174C (S2-S3), R243C (S4-S5), R366Q (proximal c-terminus) and R555C (distal c-terminus). Mutations in the c-terminus of IKs (both proximal and distal) enhanced channel sensitivity to changes in membrane PIP(2) levels, consistent with a decrease in apparent channel-PIP(2) affinity. These mutant channels were more sensitive to inhibition caused by receptor mediated PIP(2)-depletion and more sensitive to stimulation of PIP(2) production, by overexpression of phosphatidylinositol-4-phosphate-5-kinase (PI5-kinase). In addition, c-terminus mutants showed a potentiated regulation by PKA. On the other hand, for the two cytoplasmic-loop mutations, an impaired activation by PKA was observed. The effects of the mutations on PKC stimulation of the channel paralleled the effects on PKA stimulation, suggesting that both regulatory inputs are similarly affected by the mutations. We tested whether PKC-mediated activation of IKs, similarly to the PKA-mediated activation, can regulate the channel response to PIP(2). After PKC activation, channel was less sensitive to changes in membrane PIP(2) levels, consistent with an increase in apparent channel-PIP(2) affinity. PKC-activated channel was less sensitive to inhibition caused by block of synthesis of PIP(2) by the lipid kinase inhibitor wortmannin and less sensitive to stimulation of PIP(2) production. Our data indicates that stimulation by PKA and PKC can partially rescue LQT1 mutant channels with weakened response to PIP(2) by strengthening channel interactions with PIP(2).


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Ion Channel Gating , KCNQ1 Potassium Channel/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Kinase C/metabolism , Romano-Ward Syndrome/enzymology , Animals , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Genotype , Humans , KCNQ1 Potassium Channel/genetics , Membrane Potentials , Mutation , Oocytes , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/metabolism , Romano-Ward Syndrome/genetics , Romano-Ward Syndrome/physiopathology , Time Factors , Xenopus
12.
J Mol Cell Cardiol ; 46(5): 704-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19233191

ABSTRACT

KCNQ1 is co-assembled with KCNE1 subunits in the heart to form the cardiac delayed rectifier K(+) current (IKs), which is one of the main currents responsible for myocyte repolarization. The most commonly inherited form of cardiac arrhythmias, long-QT syndrome type 1 (LQT1), is due to mutations on KCNQ1. Gq-coupled receptors (GqPCRs) are known to mediate positive inotropism in human ventricular myocardium. The mechanism of IKs current modulation by GqPCRs remains incompletely understood. Here we studied the molecular mechanisms underlying Gq regulation of the IKs channel. Heterologously expressed IKs (human KCNQ1/KCNE1 subunits) was measured in Xenopus oocytes, expressed together with GqPCRs. Our data from several GqPCRs shows that IKs is regulated in a biphasic manner, showing both an activation and an inhibition phase. Receptor-mediated inhibition phase was irreversible when recycling of agonist-sensitive pools of phosphatidylinositol-4,5-bisphosphate (PIP2) was blocked by the lipid kinase inhibitor wortmannin. In addition, stimulation of PIP(2) production, by overexpression of phosphatidylinositol-4-phosphate-5-kinase (PIP5-kinase), decreased receptor-mediated inhibition. The receptor-mediated activation phase was inhibited by the PKC inhibitor calphostin C and by a mutation in a putative PKC phosphorylation site in the KCNE1 subunit. Our results indicate that the depletion of membrane PIP(2) underlies receptor-mediated inhibition of IKs and that phosphorylation by PKC of the KCNE1 subunit underlies the GqPCR-mediated channel activation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Ion Channel Gating , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels/metabolism , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/metabolism , Acetylcholine/pharmacology , Animals , Calcium/metabolism , Enzyme Activation/drug effects , Humans , Hydrolysis/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Kinetics , Models, Biological , Phosphorylation/drug effects , Xenopus
13.
Channels (Austin) ; 1(2): 124-34, 2007.
Article in English | MEDLINE | ID: mdl-18690021

ABSTRACT

Neurotransmitter and hormone regulation of cellular function can result from a concomitant stimulation of different signaling pathways. Signaling cascades are strongly regulated during disease and are often targeted by commonly used drugs. Crosstalk of different signaling pathways can have profound effects on the regulation of cell excitability. Members of all the three main structural families of potassium channels: inward-rectifiers, voltage-gated and 2-P domain, have been shown to be regulated by direct phosphorylation and Gq-coupled receptor activation. Here we test members of each of the three families, Kir3.1/Kir3.4, KCNQ1/KCNE1 and TREK-1 channels, all of which have been shown to be regulated directly by phosphatidylinositol bisphosphate (PIP2). The three channels are inhibited by activation of Gq-coupled receptors and are differentially regulated by protein kinase A (PKA). We show that Gq-coupled receptor regulation can be physiologically modulated directly through specific channel phosphorylation sites. Our results suggest that PKA phosphorylation of these channels affects Gq-coupled receptor inhibition through modulation of the channel sensitivity to PIP2.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels/physiology , Type C Phospholipases/metabolism , Acetylcholine/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Female , Fluorescence Resonance Energy Transfer , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Hydrolysis , Kidney/cytology , Microscopy, Confocal , Oocytes , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Xenopus
14.
Nat Neurosci ; 8(5): 626-34, 2005 May.
Article in English | MEDLINE | ID: mdl-15852009

ABSTRACT

The subjective feeling of cold is mediated by the activation of TRPM8 channels in thermoreceptive neurons by cold or by cooling agents such as menthol. Here, we demonstrate a central role for phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) in the activation of recombinant TRPM8 channels by both cold and menthol. Moreover, we show that Ca(2+) influx through these channels activates a Ca(2+)-sensitive phospholipase C and that the subsequent depletion of PI(4,5)P(2) limits channel activity, serving as a unique mechanism for desensitization of TRPM8 channels. Finally, we find that mutation of conserved positive residues in the highly conserved proximal C-terminal TRP domain of TRPM8 and two other family members, TRPM5 and TRPV5, reduces the sensitivity of the channels for PI(4,5)P(2) and increases inhibition by PI(4,5)P(2) depletion. These data suggest that the TRP domain of these channels may serve as a PI(4,5)P(2)-interacting site and that regulation by PI(4,5)P(2) is a common feature of members of the TRP channel family.


Subject(s)
Cold Temperature , Ion Channels/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Sensory Receptor Cells/metabolism , Thermosensing/physiology , Animals , COS Cells , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Chlorocebus aethiops , Ion Channels/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Menthol/pharmacology , Mutation/physiology , Neoplasm Proteins/genetics , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Sensory Receptor Cells/drug effects , TRPV Cation Channels , Thermosensing/drug effects
15.
J Physiol ; 564(Pt 1): 117-29, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15677683

ABSTRACT

Two-pore (2-P) domain potassium channels are implicated in the control of the resting membrane potential, hormonal secretion, and the amplitude, frequency and duration of the action potential. These channels are strongly regulated by hormones and neurotransmitters. Little is known, however, about the mechanism underlying their regulation. Here we show that phosphatidylinositol 4,5-bisphosphate (PIP2) gating underlies several aspects of 2-P channel regulation. Our results demonstrate that all four 2-P channels tested, TASK1, TASK3, TREK1 and TRAAK are activated by PIP2. We show that mechanical stimulation may promote PIP2 activation of TRAAK channels. For TREK1, TASK1 and TASK3 channels, PIP2 hydrolysis underlies inhibition by several agonists. The kinetics of inhibition by the PIP2 scavenger polylysine, and the inhibition by the phosphatidylinositol 4-kinase inhibitor wortmannin correlated with the level of agonist-induced inhibition. This finding suggests that the strength of channel PIP2 interactions determines the extent of PLC-induced inhibition. Finally, we show that PIP2 hydrolysis modulates voltage dependence of TREK1 channels and the unrelated voltage-dependent KCNQ1 channels. Our results suggest that PIP2 is a common gating molecule for K+ channel families despite their distinct structures and physiological properties.


Subject(s)
Ion Channel Gating/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Voltage-Gated/agonists , Potassium Channels, Voltage-Gated/metabolism , Androstadienes/pharmacology , Animals , Female , Humans , Hydrolysis/drug effects , Ion Channel Gating/drug effects , Mice , Polylysine/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Protein Structure, Tertiary/physiology , Rats , Wortmannin , Xenopus
16.
J Biol Chem ; 278(31): 28865-71, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12754254

ABSTRACT

Sorcin is a penta-EF hand Ca2+-binding protein that associates with both cardiac ryanodine receptors and L-type Ca2+ channels and has been implicated in the regulation of intracellular Ca2+ cycling. To better define the function of sorcin, we characterized transgenic mice in which sorcin was overexpressed in the heart. Transgenic mice developed normally with no evidence of cardiac hypertrophy and no change in expression of other calcium regulatory proteins. In vivo hemodynamics revealed significant reductions in global indices of contraction and relaxation. Contractile abnormalities were also observed in isolated adult transgenic myocytes, along with significant depression of Ca2+ transient amplitudes. Whole cell ICa density and the time course of activation were normal in transgenic myocytes, but the rate of inactivation was significantly accelerated. These effects of sorcin on L-type Ca2+ currents were confirmed in Xenopus oocyte expression studies. Finally, we examined the expression of sorcin in normal and failing hearts from spontaneous hypertensive heart failure rats. In normal myocardium, sorcin extensively co-localized with ryanodine receptors at the Z-lines, whereas in myopathic hearts the degree of co-localization was markedly disrupted. Together, these data indicate that sorcin modulates intracellular Ca2+ cycling and Ca2+ influx pathways in the heart.


Subject(s)
Calcium-Binding Proteins/physiology , Calcium/metabolism , Myocardial Contraction/physiology , Animals , Calcium Channels, L-Type/physiology , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/genetics , Cardiomyopathies/metabolism , Gene Expression , Humans , Immunohistochemistry , Kinetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/chemistry , Myocardium/metabolism , Oocytes/metabolism , Ryanodine Receptor Calcium Release Channel/analysis , Transfection , Xenopus laevis
17.
Neuron ; 37(6): 963-75, 2003 Mar 27.
Article in English | MEDLINE | ID: mdl-12670425

ABSTRACT

KCNQ channels belong to a family of potassium ion channels with crucial roles in physiology and disease. Heteromers of KCNQ2/3 subunits constitute the neuronal M channels. Inhibition of M currents, by pathways that stimulate phospholipase C activity, controls excitability throughout the nervous system. Here we show that a common feature of all KCNQ channels is their activation by the signaling membrane phospholipid phosphatidylinositol-bis-phosphate (PIP(2)). We show that wortmannin, at concentrations that prevent recovery from receptor-mediated inhibition of M currents, blocks PIP(2) replenishment to the cell surface. Moreover, we identify a C-terminal histidine residue, immediately proximal to the plasma membrane, mutation of which renders M channels less sensitive to PIP(2) and more sensitive to receptor-mediated inhibition. Finally, native or recombinant channels inhibited by muscarinic agonists can be activated by PIP(2). Our data strongly suggest that PIP(2) acts as a membrane-diffusible second messenger to regulate directly the activity of KCNQ currents.


Subject(s)
Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels/physiology , Receptors, Muscarinic/physiology , Androstadienes/pharmacology , Animals , Bradykinin/pharmacology , CHO Cells , Cells, Cultured , Cricetinae , Electric Conductivity , Enzyme Inhibitors/pharmacology , Female , Hydrolysis , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , Microscopy, Confocal , Mutation , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Channels/genetics , Potassium Channels, Voltage-Gated , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1 , Recombinant Proteins , Second Messenger Systems , Signal Transduction , Superior Cervical Ganglion/cytology , Tetraethylammonium/pharmacology , Transfection , Wortmannin , Xenopus laevis
18.
Proc Natl Acad Sci U S A ; 100(2): 745-50, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12525701

ABSTRACT

Phosphoinositides are critical regulators of ion channel and transporter activity. Defects in interactions of inwardly rectifying potassium (Kir) channels with phosphoinositides lead to disease. ATP-sensitive K(+) channels (K(ATP)) are unique among Kir channels in that they serve as metabolic sensors, inhibited by ATP while stimulated by long-chain (LC) acyl-CoA. Here we show that K(ATP) are the least specific Kir channels in their activation by phosphoinositides and we demonstrate that LC acyl-CoA activation of these channels depends on their low phosphoinositide specificity. We provide a systematic characterization of phosphoinositide specificity of the entire Kir channel family expressed in Xenopus oocytes and identify molecular determinants of such specificity. We show that mutations in the Kir2.1 channel decreasing phosphoinositide specificity allow activation by LC acyl-CoA. Our data demonstrate that differences in phosphoinositide specificity determine the modulation of Kir channel activity by distinct regulatory lipids.


Subject(s)
Phosphatidylinositols/pharmacology , Potassium Channels/drug effects , Acyl Coenzyme A/pharmacology , Amino Acid Sequence , Animals , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , Molecular Sequence Data , Potassium Channels/chemistry , Potassium Channels/physiology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Xenopus
19.
Neuron ; 34(6): 933-44, 2002 Jun 13.
Article in English | MEDLINE | ID: mdl-12086641

ABSTRACT

Inwardly rectifying K(+) (Kir) channels are important regulators of resting membrane potential and cell excitability. The activity of Kir channels is critically dependent on the integrity of channel interactions with phosphatidylinositol 4,5-bisphosphate (PIP(2)). Here we identify and characterize channel-PIP(2) interactions that are conserved among Kir family members. We find basic residues that interact with PIP(2), two of which have been associated with Andersen's and Bartter's syndromes. We show that several naturally occurring mutants decrease channel-PIP(2) interactions, leading to disease.


Subject(s)
Conserved Sequence/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Amino Acid Sequence/genetics , Animals , Bartter Syndrome/genetics , Bartter Syndrome/metabolism , Female , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/metabolism , Ion Channel Gating/genetics , Membrane Potentials/genetics , Molecular Sequence Data , Mutation/genetics , Oocytes/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...