Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Respir Physiol Neurobiol ; 154(3): 356-71, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16466972

ABSTRACT

This study examines developmental changes in CB glomus cell depolarization, intracellular calcium ([Ca(2+)](i)) and the magnitude of an O(2)-sensitive background ionic conductance that may play roles in the postnatal increase in oxygen sensitivity of glomus cells isolated from rats of 1-3 days and 11-14 days postnatal age. Using fura-2 and perforated patch whole cell recordings, we simultaneously measured [Ca(2+)](i) and membrane potential (E(m)) during normoxia and hypoxia. Resting E(m) in normoxia was similar at both ages. Hypoxia caused a larger E(m) depolarization and correspondingly larger [Ca(2+)](i) response in glomus cells from 11- to 14-day-old rats compared to 1-3-day-old rats. E(m) and [Ca(2+)](i) responses to 40mM K(+) were identical between the two age groups. Under normoxic conditions both age groups had similar background conductances. Under anoxic conditions (at resting membrane potential) background K(+) conductance decreased significantly more in cells from 11- to 14-day-old rats compared to cells from 1- to 3-day-old rats. Glomus cells from newborns therefore have less O(2)-sensitive background K(+) conductance. These results support the hypothesis that postnatal maturation of glomus cell O(2) sensitivity involves developmental regulation of the expression and/or O(2)-sensitivity of background ionic conductances.


Subject(s)
Aging , Animals, Newborn/growth & development , Carotid Body/physiopathology , Hypoxia/physiopathology , 4-Aminopyridine/pharmacology , Animals , Animals, Newborn/metabolism , Calcium/metabolism , Carotid Body/metabolism , Carotid Body/pathology , Electric Capacitance , Electric Impedance , Hypoxia/metabolism , Hypoxia/pathology , Intracellular Membranes/metabolism , Membrane Potentials , Membranes/metabolism , Membranes/physiopathology , Osmolar Concentration , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Rats , Rest , Tetraethylammonium/pharmacology
2.
J Biol Chem ; 276(42): 39053-9, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11489900

ABSTRACT

Calcium sensing (CaR) and Group I metabotropic glutamate receptors exhibit overlapping expression patterns in brain, and share common signal transduction pathways. To determine whether CaR and Group I metabotropic glutamate receptors (mGluRs) (mGluR1alpha and mGluR5) can form heterodimers, we immunoprecipitated CaR from bovine brain and observed co-precipitation of mGluR1alpha. CaR and mGluR1alpha co-localize in hippocampal and cerebellar neurons, but are expressed separately in other brain regions. In vitro transfection studies in HEK-293 cells established the specificity and disulfide-linked nature of the CaR:mGluR1alpha (CaR:mGluR5) interactions. CaR:mGluR1alpha (CaR:mGluR5) heterodimers exhibit altered trafficking via Homer 1c when compared with CaR:CaR homodimers. CaR becomes sensitive to glutamate-mediated internalization when present in CaR:mGluR1alpha heterodimers. These results demonstrate cross-family covalent heterodimerization of CaR with Group I mGluRs, and increase the potential role(s) for CaR in modulating neuronal function.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/chemistry , Animals , Brain/metabolism , Cattle , Cell Line , Cerebellum/metabolism , Dimerization , Disulfides , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , Humans , Microscopy, Fluorescence , Neurons/chemistry , Precipitin Tests , Protein Binding , Rats , Transfection
3.
Am J Physiol Cell Physiol ; 280(6): C1412-21, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11350736

ABSTRACT

Parathyroid hormone secretion is exquisitely sensitive to small changes in serum Ca2+ concentration, and these responses are transduced via the Ca2+-sensing receptor (CaR). We utilized heterologous expression in HEK-293 cells to determine the effects of small, physiologically relevant perturbations in extracellular Ca2+ on CaR signaling via phosphatidylinositol-phospholipase C, using changes in fura 2 fluorescence to quantify intracellular Ca2+. Chronic exposure of CaR-transfected cells to Ca2+ in the range from 0.5 to 3 mM modulated the resting intracellular Ca2+ concentration and the subsequent cellular responses to acute extracellular Ca2+ perturbations but had no effect on thapsigargin-sensitive Ca2+ stores. Modest, physiologically relevant increases in extracellular Ca2+ concentration (0.5 mM increments) caused sustained (30-40 min) low-frequency oscillations of intracellular Ca2+ (approximately 45 s peak to peak interval). Oscillations were eliminated by 1 microM thapsigargin but were insensitive to protein kinase inhibitors (staurosporine, KN-93, or bisindolylmaleimide I). Staurosporine did increase the fraction of cells oscillating at a given extracellular Ca2+ concentration. Serum Ca2+ concentrations thus chronically regulate cells expressing CaR, and small perturbations in extracellular Ca2+ alter both resting intracellular Ca2+ as well as Ca2+ dynamics.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Receptors, Cell Surface/metabolism , Cell Line , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Fura-2 , Gene Expression/physiology , Humans , Kidney/cytology , Protein Kinase Inhibitors , Protein Kinases/metabolism , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics , Thapsigargin/pharmacology , Transfection
5.
J Biol Chem ; 274(17): 11629-34, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10206973

ABSTRACT

Calcium-sensing receptors are present in membranes as dimers that can be reduced to monomers with sufhydryl reagents. All studies were carried out on the human calcium-sensing receptor tagged at the carboxyl terminus with green fluorescent protein (hCaR-GFP) to permit identification and localization of expressed proteins. Truncations containing either the extracellular agonist binding domain plus transmembrane helix 1 (ECD/TMH1-GFP) or the transmembrane domain plus the intracellular carboxyl terminus (TMD/carboxyl terminus-GFP) were used to identify the dimerization domain. ECD/TMH1-GFP was a dimer in the absence of reducing reagents, whereas TMD/carboxyl-terminal GFP was a monomer in the absence or presence of reducing agents, suggesting that dimerization occurs via the ECD. To identify the residue(s) involved in dimerization within the ECD, cysteine --> serine point mutations were made in residues that are conserved between hCaR and metabotropic glutamate receptors. Mutations at positions 60 and 131 were expressed at levels comparable to wild type in HEK 293 cells, had minimal effects on hCaR function, and did not eliminate dimerization, whereas mutations at positions 101 and 236 greatly decreased receptor expression and resulted in significant amounts of monomer in the absence of reducing agents. The double point mutant hCaR(C101S/C236S)-GFP was expressed more robustly than either C101S or C236S and covalent dimerization was eliminated. hCaR(C101S/C236S)-GFP had a decreased affinity for extracellular Ca2+ and slower response kinetics upon increases or decreases in agonist concentration. These results suggest that covalent, disulfide bond-mediated dimerization of the calcium-sensing receptor contributes to stabilization of the ECD and to acceleration of the transitions between inactive and active receptor conformations.


Subject(s)
Cysteine/metabolism , Receptors, Cell Surface/metabolism , Serine/metabolism , Base Sequence , Cell Line , Cysteine/genetics , DNA Primers , Dimerization , Disulfides/chemistry , Disulfides/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Point Mutation , Receptors, Calcium-Sensing , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Serine/genetics
6.
Biochim Biophys Acta ; 1416(1-2): 195-207, 1999 Jan 12.
Article in English | MEDLINE | ID: mdl-9889364

ABSTRACT

Activation of the influx mode of the Na+,K+,Cl- cotransporter (NKCC) by extracellular Na+, K+ and Cl- was studied using the internally dialyzed squid giant axon. Cooperative interactions among the three transported ions were assessed using ion activation of NKCC-mediated 36Cl influx under two sets of experimental conditions. The first, or control condition, used high, non-limiting concentrations of two of the cotransported ions (the co-ions) while activating cotransport with the third ion. Under this non-limiting co-ion condition the calculated Vmax of the cotransporter was between 57 and 60 pmol/cm2/s. The apparent activation (KApp, or half-saturation) constants were: K+, 9 mM; Na+, 52 mM; and Cl-, 146 mM. The second condition used limiting co-ion concentration conditions. In this case, activation by each ion was determined when one of the other two co-ions was present at or near its apparent half-saturation concentration as determined above. Under these limiting conditions, the KApp values for all three co-ions were significantly increased regardless of which co-ion was present at a limiting concentration. The effects on the apparent Vmax were more complicated. When K+ was the limiting co-ion, there was little effect on the Vmax for Na+ or Cl- activation. In contrast, limiting concentrations of Na+ or Cl- both resulted in a large reduction of the apparent Vmax when activating with the other two co-ions. These results are consistent with an ordered binding mechanism for the NKCC in which K+ binds before Na+ or Cl-. Physiological implications for these results are discussed.


Subject(s)
Axons/metabolism , Carrier Proteins/metabolism , Animals , Chlorides/metabolism , Chlorides/pharmacology , Decapodiformes , Osmolar Concentration , Potassium/metabolism , Potassium/pharmacology , Sodium/metabolism , Sodium/pharmacology , Sodium-Potassium-Chloride Symporters
7.
J Biol Chem ; 273(45): 29712-8, 1998 Nov 06.
Article in English | MEDLINE | ID: mdl-9792684

ABSTRACT

Calcium sensing receptors are part of a growing G protein-coupled receptor family, which includes metabotropic glutamate, gamma-aminoisobutyric acid, and pheromone receptors. The distinctive structural features of this family include large extracellular domains that bind agonist and large intracellular, carboxyl-terminal domains of as yet undefined function(s). We have explored the contribution(s) of the carboxyl terminus of the human calcium sensing receptor (CaR) by assessing extracellular Ca2+-mediated changes in intracellular Ca2+ in individual HEK-293 cells transfected with CaR clones. In-frame fusion of EGFP to the carboxyl terminus of CaR had no effect on either the dose response for extracellular Ca2+ activation or CaR desensitization. Carboxyl-terminal truncations, fused in-frame with EGFP (CaRDelta1024-EGFP, CaRDelta908-EGFP, CaRDelta886-EGFP, and CaRDelta868-EGFP), were assessed for alterations in Ca2+-dependent activation or desensitization. Significant effects on the dose-response relation for extracellular Ca2+ were observed only for the CaRDelta868 truncation, which exhibited a decreased affinity for extracellular Ca2+ and a decrease in the apparent cooperativity for Ca2+-dependent activation. The alterations in extracellular Ca2+ affinity and cooperativity observed with CaRDelta868 were recapitulated by a point mutation, T876D, in the full-length CaR-EGFP background. All truncations with wild type dose-response relations exhibited desensitization time courses that were comparable to the full-length CaR, whereas the CaRDelta868 receptor desensitized completely after two exposures to 10 mM Ca2+. Interestingly, the CaR point mutation T876D exhibited desensitization comparable to wild type CaR, suggesting that this mutation specifically modifies CaR cooperativity. In conclusion, these studies suggest that amino acid residues between 868 and 886 are critical to the apparent cooperativity of Ca2+-mediated activation of G proteins and to CaR desensitization.


Subject(s)
Calcium/metabolism , Extracellular Space/metabolism , Receptors, Cell Surface/metabolism , Base Sequence , Cell Line , Cell Membrane/metabolism , DNA Primers , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Point Mutation , Protein Conformation , Receptors, Calcium-Sensing , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/metabolism
8.
Am J Physiol ; 273(4): C1168-75, 1997 10.
Article in English | MEDLINE | ID: mdl-9357760

ABSTRACT

Expression of Ca2+-sensing receptors (CaR) was demonstrated in several human intestinal epithelial cell lines (T84, HT-29, and Caco-2) and in rat intestinal epithelium by both reverse transcriptase-polymerase chain reaction (PCR) and Northern blotting of RNA. Restriction patterns of the PCR products were of the sizes predicted by the human and rat sequences. CaR agonists (Ca2+, poly-L-arginine, protamine) mediated an increase in intracellular Ca2+ in HT-29-18-C1 cells (monitored by changes in fura 2 fluorescence), which was dependent on release from thapsigargin-sensitive stores. U-73122, an inhibitor of phosphatidylinositol-phospholipase C, eliminated the CaR agonist-mediated rise in intracellular Ca2+, whereas its inactive analog, U-73343, had no effect. Pertussis toxin pretreatment had no effect on CaR agonist-mediated modulation of intracellular Ca2+. Taken together, these studies demonstrate that CaR are expressed in intestinal epithelial cells and couple to mobilization of intracellular Ca2+. The presence of CaR in intestinal epithelial cells presents a new locus for investigations into the role(s) of extracellular Ca2+ in modulating intestinal epithelial cell differentiation and transepithelial Ca2+ transport.


Subject(s)
Intestinal Mucosa/metabolism , Receptors, Cell Surface/biosynthesis , Transcription, Genetic , Animals , Calcium/pharmacology , Cecum , Cell Line , Colon , DNA Primers , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Exons , Humans , Intestine, Small , Introns , Kinetics , Neurotensin/pharmacology , Peptides/pharmacology , Phosphatidylinositol Diacylglycerol-Lyase , Polymerase Chain Reaction , Pyrrolidinones/pharmacology , Rats , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics , Thapsigargin/pharmacology , Transcription, Genetic/drug effects , Tumor Cells, Cultured , Type C Phospholipases/antagonists & inhibitors
9.
Am J Physiol ; 272(3 Pt 1): C957-65, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9124532

ABSTRACT

Expression of the human gene A4 is enriched in the colonic epithelium and is transcriptionally activated on differentiation of colonic epithelial cells in vitro (M. M. Oliva, T. C. Wu, and V. W. Yang. Arch. Biochem. Biophys. 302: 183-192, 1993). A4 cDNA contains an open reading frame that predicts a polypeptide of 17 kDa. To determine the function of the A4 protein, we characterized its biochemical and physiological properties. Hydropathy analysis of deduced A4 amino acid sequence revealed four putative membrane-spanning alpha-helices. The hydrophobic nature of A4 was confirmed by its being extractable with organic solvents. Immunocytochemical studies of cells expressing A4 localized it to the endoplasmic reticulum. Moreover, A4 multimerized in vivo as determined by coimmunoprecipitation experiments. The four-transmembrane topology and biophysical characteristics of A4 suggest that it belongs to a family of integral membrane proteins called proteolipids, some of which multimerize to form ion channels. Subsequent electrophysiological studies of nuclei isolated from microinjected Xenopus laevis oocytes transiently expressing A4 showed the appearance of a 28-pS channel. Thus our studies indicate that A4 is a colonic epithelium-enriched protein localized to the endoplasmic reticulum and that, similar to other proteolipids, A4 multimerizes and exhibits characteristics of an ion channel.


Subject(s)
Intestinal Mucosa/physiology , Ion Channels/physiology , Membrane Proteins/physiology , Proteolipids/physiology , Amino Acid Sequence , Animals , Blotting, Western , Cell Compartmentation , Endoplasmic Reticulum/metabolism , Epithelium/physiology , Humans , Intracellular Membranes/metabolism , MARVEL Domain-Containing Proteins , Macromolecular Substances , Membrane Potentials , Membrane Proteins/chemistry , Molecular Sequence Data , Precipitin Tests , Recombinant Proteins , Solubility , Tumor Cells, Cultured , Xenopus laevis
10.
J Gen Physiol ; 109(2): 245-53, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9041452

ABSTRACT

Receptor-mediated activation of heterotrimeric G proteins leading to dissociation of the G alpha subunit from G beta gamma is a highly conserved signaling strategy used by numerous extracellular stimuli. Although G beta gamma subunits regulate a variety of effectors, including kinases, cyclases, phospholipases, and ion channels (Clapham, D.E., and E.J. Neer. 1993. Nature (Lond.). 365:403-406), few tools exist for probing instantaneous G beta gamma-effector interactions and little is known about the kinetic contributions of effectors to the signaling process. In this study, we used the atrial muscarinic K + channel, which is activated by direct interactions with G beta gamma subunits (Logothetis, D.E., Y. Kurachi J. Galper, E.J. Neer, and D.E. Clap. 1987. Nature (Lond.). 325:321-326; Wickman, K., J. A. Iniguez-Liuhi, P.A. Davenport, R. Taussig, G.B. Krapivinsky, M.E. Linder, A.G. Gilman, and D.E. Clapham. 1994. Nature (Lond.). 366: 654-663; Huang, C.-L., P.A. Slesinger, P.J. Casey, Y.N. Jan, and L.Y. Jan. 1995. Neuron. 15:1133-1143), as a sensitive reporter of the dynamics of G beta gamma-effector interactions. Muscarinic K+ channels exhibit bursting behavior upon G protein activation, shifting between three distinct functional modes, characterized by the frequency of channel openings during individual bursts. Acetylcholine concentration (and by inference, the concentration of activated G beta gamma) controls the fraction of time spent in each mode without changing either the burst duration or channel gating within individual modes. The picture which emerges is of a G beta gamma effector with allosteric regulation and an intrinsic "off" switch which serves to limit its own activation. These two features combine to establish exquisite channel sensitivity to changes in G beta gamma concentration, and may be indicative of the factors regulating other G beta gamma-modulated effectors.


Subject(s)
GTP-Binding Proteins/metabolism , Ion Channel Gating/physiology , Potassium Channels/physiology , Receptors, Muscarinic/metabolism , Signal Transduction/physiology , Animals , Electrophysiology , In Vitro Techniques , Kinetics , Membrane Potentials/physiology , Myocardium/cytology , Myocardium/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Rana catesbeiana
12.
J Gen Physiol ; 107(2): 261-70, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8833345

ABSTRACT

Bumetanide-sensitive (BS) unidirectional fluxes of (36)Cl- or (22)Na+ were measured in internally dialyzed squid giant axons while varying the intra- or extracellular concentrations of Na+ and/or Cl-. Raising either [Cl-]i or [Na+]i resulted in a concentration-dependent reduction of the BS influx of both (36)Cl- and (22)Na+. Raising [Cl-]i above 200 mM completely blocked BS influxes. However, raising [Na+]i to 290 mM resulted in saturable but incomplete inhibition of both BS Na+ influx and BS Cl- influx. The consequences of varying intracellular Cl- on cotransporter effluxes were complex. At lower [Cl-]i values (below 100 mM) intracellular Cl- activated cotransporter effluxes. Surprisingly, however, raising [Cl-]i levels > 125 mM resulted in a [Cl-]i-dependent inhibition of BS effluxes of both Na+ and Cl-. On the other hand, raising [Na+]i resulted only in the activation of the BS Na+ efflux; intracellular Na+ did not inhibit BS efflux even at 290 mM. The inhibitory effects of intracellular Na+ on cotransporter-mediated influxes, and lack of inhibitory effects on BS effluxes, are consistent with the trans-side inhibition expected for an ordered binding/release model of cotransporter operation. However, the inhibitory effects of intracellular Cl- on both influxes and effluxes are not explained by such a model. These data suggest that Cl may interact with an intracellular site (or sites), which does not mediate Cl transport, but does modulate the transport activity of the Na+, K+, Cl- cotransporter.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Chlorides/metabolism , Sodium/metabolism , Animals , Axons/chemistry , Bumetanide/pharmacology , Carrier Proteins/drug effects , Chlorides/pharmacology , Decapodiformes , Diuretics/pharmacology , Membrane Proteins/metabolism , Potassium/metabolism , Sodium/pharmacology , Sodium-Potassium-Chloride Symporters
13.
Genome Res ; 5(5): 453-63, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8808466

ABSTRACT

A mutation in the GIRK2 inwardly rectifying K+ channel was mapped recently to the region of mouse chromosome 16 containing the wv gene and shown to occur in mutant but not in wild-type mice. We demonstrate tight linkage of the Girk2 mutation to the wv phenotype and refine the localization of the weaver (wv) gene on recombinational and physical maps. This linkage between Girk2 and wv has existed since at least 1988 in descendants of the original mutation maintained in C57BL/6 animals. Girk2 is shown to be transcribed in brain before the first recognized manifestation of the wv phenotype and in cultures of granule cells (GCs) isolated from cerebellum at postnatal day 8. Wild-type GCs grown in this culture system display an important developmental property--the ability to extend neurites. However, no inwardly rectifying K+ current is detected in GCs cultured from either wv/wv or +/+ cerebellum under a variety of conditions that activate related channels in other tissues. This suggests that if the Girk2 mutation is responsible for the wv phenotype, it does not act by altering these electrical properties of developing GCs.


Subject(s)
Mice, Neurologic Mutants/genetics , Mutation , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Animals , Base Sequence , Cells, Cultured , Cerebellum/metabolism , Cerebellum/pathology , Chromosome Mapping , Chromosomes, Artificial, Yeast , Crosses, Genetic , DNA Primers/genetics , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Genetic Linkage , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype , Polymerase Chain Reaction , Recombination, Genetic
14.
Am J Physiol ; 269(4 Pt 1): C878-83, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7485456

ABSTRACT

The Na(+)-K(+)-Cl- cotransporter of the squid giant axon requires ATP and is inhibited by intracellular Cl- (Cli-) in a concentration-dependent manner ([Cl-]i > or = 200 mM completely inhibits the cotransporter). In the present study we address the question of whether inhibition of cotransport by Cli- is due to a Cl(i-)-induced increase of protein phosphatase activity. Intracellular dialysis was used to apply the phosphatase inhibitor okadaic acid (OKA) under conditions of [Cl-]i at 0, 150, or 300 mM during measurement of cotransporter-mediated unidirectional Cl- influx into axons. At 0 mM [Cl-]i, the application of 250 nM OKA had no effect on the cotransport-mediated Cl- influx when axons were dialyzed with the normal intracellular ATP concentration ([ATP]i = 4 mM). Reduction of [ATP] to 50 microM resulted in a significant decrease of the bumetanide-sensitive CL- influx, which could be partially reversed by OKA treatment. Similarly, in ATP-limited axons with [Cl-]i at 150 mM, cotransporter influx was partially stimulated by treatment with OKA. However, axons dialyzed with 300 mM [Cl-]i ([ATP]i = 50 microM) had no measurable cotransport influx, nor was subsequent treatment with OKA able to induce a cotransport-mediated Cl- influx. We conclude that the inhibition of cotransport caused by Cli- is not the result of an increase in the OKA-sensitive protein phosphatase activity.


Subject(s)
Carrier Proteins/metabolism , Chlorides/metabolism , Ethers, Cyclic/pharmacology , Adenosine Triphosphate/metabolism , Animals , Axons/metabolism , Decapodiformes , Intracellular Membranes/metabolism , Okadaic Acid , Osmolar Concentration , Phosphoprotein Phosphatases/antagonists & inhibitors , Sodium-Potassium-Chloride Symporters
15.
Am J Physiol ; 266(4 Pt 1): C1128-32, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8178960

ABSTRACT

Acetylcholine (ACh) binding to atrial muscarinic receptors activates an inwardly rectifying K+ current (IK[ACh]) via a pertussis toxin-sensitive GTP-binding protein (GK). The muscarinic K+ channel (termed GIRK1) has been cloned, and the nucleotide sequence contains nine consensus sites for protein kinase C (PKC) phosphorylation (16). Dephosphorylation of the muscarinic K+ channel has been implicated in rapid IK[ACh] desensitization in the presence of agonist (13). Staurosporine is a widely used membrane-permeant inhibitor of PKC and other protein kinases (7), including G protein-coupled receptor kinases. We investigated the role of phosphorylation in the regulation of IK[ACh] by examining the effect of a variety of protein kinase inhibitors. Staurosporine produced a rapid and reversible dose-dependent decrease in IK[ACh], activated by either GTP or guanosine 5'-O-(3-thiotriphosphate) (GTP gamma S). Other PKC inhibitors, including calphostin C and K-252b, were without effect on GTP gamma S-activated IK[ACh]. In excised patches of atrial membrane under nonphosphorylating conditions (0 ATP, 1 mM 5'-adenylylimidodiphosphate), staurosporine reversibly reduced muscarinic K+ channel activity without altering single-channel current amplitude. These results suggest that staurosporine inhibits IK[ACh] by a mechanism independent of intracellular protein kinases.


Subject(s)
Alkaloids/pharmacology , Myocardium/metabolism , Potassium Channel Blockers , Protein Kinases/physiology , Receptors, Muscarinic/metabolism , Adenylyl Imidodiphosphate/pharmacology , Animals , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Heart Atria , Myocardium/cytology , Protein Kinase Inhibitors , Rana catesbeiana , Staurosporine
16.
J Gen Physiol ; 102(1): 125-41, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8376954

ABSTRACT

The effects of leukotriene C4 (LTC4) on activation of muscarinic acetylcholine receptor (mAChR)-stimulated, inwardly rectifying K+ current (IK[ACh]) were examined in single bullfrog atrial myocytes using the whole-cell patch clamp technique. LTC4 produced a reversible, concentration-dependent increase in steady-state, guanosine-gamma-thiotriphosphate (GTP gamma S)-activated IK[ACh], with a K0.5 of 3.1 microM. LTC4 also increased the rate of GTP gamma S-mediated IK[ACh] activation, both in the absence and presence of 1 nM ACh, with comparable K0.5 values of 4.7 microM under basal conditions and 4.9 microM in the presence of 1 nM ACh. LTC4 did not alter the relative affinities of the G protein, Gk, for GTP gamma S and GTP. We hypothesize that all of the effects of LTC4 on the kinetics of Gk-mediated IK[ACh] activation are produced at a common site with a K0.5 of 3-5 microM. The effects of LTC4 on IK[ACh] activation are fully reversible in the presence of GTP gamma S. Under physiological conditions (i.e., intracellular GTP), 10 microM LTC4 increased the ACh-activated peak IK[ACh]. Inhibitors of cellular LTC4 production, including 5,8,11,14-eicosatetraynoic acid, baicalein, cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate, and alpha-pentyl-4-(2-quinolinylmethoxy)-benzene methanol, greatly attenuated ACh-dependent IK[ACh] activation, preventing activation of peak, and producing a lower steady-state IK[ACh] (when compared with the control response in the same cell). Addition of exogenous LTC4 was able to overcome the effects of LTC4 synthesis inhibitors, restoring both the peak and steady-state IK[ACh] responses. Although the mechanism of LTC4-mediated modulation of IK[ACh] activation is not known, our results suggest that endogenously produced lipoxygenase metabolites of arachidonic acid, specifically LTC4, are involved in the physiological process of IK[ACh] activation.


Subject(s)
Myocardium/metabolism , Potassium Channels/metabolism , Receptors, Muscarinic/drug effects , SRS-A/pharmacology , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Animals , Biotransformation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Electrophysiology , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , In Vitro Techniques , Lipoxygenase Inhibitors/pharmacology , Myocardium/cytology , Myocardium/enzymology , Potassium Channels/drug effects , Rana catesbeiana , SRS-A/antagonists & inhibitors
17.
Circ Res ; 72(4): 786-94, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8383014

ABSTRACT

Platelet-activating factor (PAF), a potent signaling lipid implicated as a mediator of pathological responses, has both negative chronotropic and inotropic effects on the heart, although the mechanism(s) involved is not well defined. Because activation of the muscarinic acetylcholine-activated K+ current (IK(ACh)) also produces a negative chronotropic and inotropic response in myocardium, this study examines whether PAF has effects on IK(ACh) in isolated bullfrog atrial myocytes under whole-cell voltage-clamp conditions. We find that 2 microM PAF increases the rate of GTP-gamma-S-mediated IK(ACh) activation (from 0.30 +/- 0.01 min-1 [n = 20] to 0.73 +/- 0.07 min-1 [n = 12], p < 0.005, in the absence of acetylcholine). This effect of 2 microM PAF was blocked by the PAF antagonist CV-3988 (5 microM, 0.33 +/- 0.14 min-1 [n = 12]), suggesting the presence of specific PAF receptors coupled to IK(ACh) activation. Further support for mediation by specific G protein-coupled PAF receptors derives from the inability of PAF to modulate IK(ACh) after maximal activation in the presence of GTP-gamma-S. Eicosatetraynoic acid (ETYA, an inhibitor of 5- and 12-lipoxygenases) did not prevent the PAF-mediated increase in the rate of IK(ACh) activation (10 microM ETYA, 0.28 +/- 0.03 min-1 [n = 7]; 10 microM ETYA plus 2 microM PAF, 0.58 +/- 0.13 min-1 [n = 8]; p < 0.05), suggesting that the observed PAF effect is not mediated by increases in arachidonic acid metabolism.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Atrial Function , Muscarine/metabolism , Myocardium/cytology , Platelet Membrane Glycoproteins , Potassium/physiology , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Acetylcholine/pharmacology , Animals , Electric Conductivity , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Lipid Metabolism , Platelet Activating Factor/pharmacology , Rana catesbeiana
18.
Hypertension ; 17(5): 684-92, 1991 May.
Article in English | MEDLINE | ID: mdl-1708743

ABSTRACT

Guanine nucleotide binding proteins couple a wide variety of receptors to ion channels via both "direct" or membrane-delimited and "indirect" second messenger-mediated pathways. This tutorial summarizes current approaches to defining the mechanisms of guanine nucleotide binding protein-mediated ion channel activation. Two well-characterized ion channels in the heart, namely, the beta-adrenergic receptor-activated calcium channel and the muscarinic receptor-activated potassium channel, are used to illustrate the criteria that can distinguish between direct and indirect guanine nucleotide binding protein-transduced pathways.


Subject(s)
GTP-Binding Proteins/physiology , Ion Channels/physiology , Animals , Calcium Channels/physiology , Humans , Myocardium/metabolism , Potassium Channels/physiology , Receptors, Adrenergic, beta/physiology , Receptors, Muscarinic/physiology
19.
J Gen Physiol ; 96(4): 735-55, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2124257

ABSTRACT

The muscarinic acetylcholine receptor (mAChR)-stimulated, inwardly rectifying K+ current (IK [ACh]) was examined in single bullfrog atrial cells using the whole-cell patch clamp technique. IK[ACh] was activated either by bath addition of 1 microM ACh or via activation of the G protein, Gk, with guanosine-gamma-thiotriphosphate (GTP gamma S). Arachidonic acid (AA) modulated IK[ACh] under both conditions. AA decreased mAChR-stimulated IK[ACh] and increased the rate of decay from the peak current (desensitization). In addition, AA affected GTP gamma S-activated IK[ACh] by modulation of Gk. The effects of AA and its metabolites on Gk were assessed by examining their effects on both the basal rate of Gk activation by GTP gamma S, and the mAChR-mediated increase in activation rate produced by nanomolar ACh. AA increased the basal rate of GTP gamma S-mediated IK[ACh] activation, but reduced the ACh-induced augmentation of this rate. All of the effects of AA on GTP gamma S-mediated IK[ACh] activation were produced by metabolites. A lipoxygenase inhibitor, nordihydroguaiaretic acid (NDGA), decreased the basal and ACh-enhanced rate of IK[ACh] activation in both the presence and absence of exogenous AA. In contrast, indomethacin (INDO), a cyclooxygenase inhibitor, increased the basal rate of IK[ACh] activation by GTP gamma S in both the presence and absence of exogenous AA, and reversed the effects of AA on the ACh-augmented basal rate. AA metabolites produced via lipoxygenase and cyclooxygenase pathways thus have opposing effects on the signal transduction pathway from mAChR to IK[ACh]. We directly tested a lipoxygenase pathway metabolite, LTC4, on GTP gamma S-mediated IK[ACh] activation and found that it not only overcame the inhibitory effects of NDGA, but also increased both the basal and ACh-augmented rate of IK[ACh] activation. From these data, we propose that AA metabolites modulate the function of Gk by altering its kinetic properties.


Subject(s)
Arachidonic Acids/pharmacology , GTP-Binding Proteins/metabolism , Myocardium/metabolism , Acetylcholine/pharmacology , Animals , Arachidonic Acid , Arachidonic Acids/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , In Vitro Techniques , Kinetics , Potassium Channels/drug effects , Potassium Channels/metabolism , Rana catesbeiana , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Am J Physiol ; 258(4 Pt 1): C749-53, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2333955

ABSTRACT

The effects of increasing extracellular osmolality on unidirectional Cl- fluxes through the Na(+)-K(+)-Cl- cotransporter were studied in internally dialyzed squid giant axons. Hyperosmotic seawater stimulated bumetanide-sensitive Cl-influx at 150 mM intracellular Cl- concentration ([Cl-]i), whereas Cl- efflux was unaffected under comparable ionic conditions. Stimulation of bumetanide-sensitive Cl- influx was proportional to the increase in extracellular osmolality. Bumetanide-sensitive Cl- influx began to increase after a latency of approximately 20 min after a stepwise increase of extracellular osmolality and continued to increase for at least 70 min. The increased bumetanide-sensitive Cl- influx measured after 65 min of exposure to hyperosmotic external fluid was a function of the intracellular Cl- concentration; stimulation by hyperosmotic external fluids was observed at physiological [Cl-]i levels (greater than 100 mM) but not at lower [Cl-]i levels. Under both normo- and hyperosmotic conditions, intracellular Cl- inhibited Na(+)-K(+)-Cl- cotransport influx in a concentration-dependent manner. However, in hyperosmotic seawater, the dose dependence of inhibition by intracellular Cl- was shifted to higher [Cl-]i values. Therefore, we conclude that hyperosmotic extracellular fluids stimulate influx via the Na(+)-K(+)-Cl- cotransport by resetting the relation between [Cl-]i and transport activity.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Chlorides/pharmacology , Animals , Axons/drug effects , Bumetanide/pharmacology , Chlorides/metabolism , Decapodiformes , In Vitro Techniques , Kinetics , Osmolar Concentration , Potassium/metabolism , Seawater , Sodium/metabolism , Sodium-Potassium-Chloride Symporters
SELECTION OF CITATIONS
SEARCH DETAIL
...