Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Neuroscience ; 122(3): 659-68, 2003.
Article in English | MEDLINE | ID: mdl-14622909

ABSTRACT

Although much progress has been made in understanding synapse formation, little is known about the mechanisms underlying synaptic maintenance and loss. The formation of agrin-induced AChR clusters on cultured myotubes requires both activation of the receptor tyrosine kinase MuSK and intracellular calcium fluxes. Here, we provide evidence that such AChR clusters are maintained by agrin/MuSK-induced intracellular calcium fluxes. Clamping intracellular calcium fluxes after AChR clusters have formed leads to rapid MuSK and AChR tyrosine dephosphorylation and cluster dispersal, even in the continued presence of agrin. Both the dephosphorylation and the dispersal are inhibited by the tyrosine phosphatase inhibitor pervanadate. In contrast, clamping intracellular calcium at the time of initial agrin stimulation has no effect on agrin-induced MuSK or AChR phosphorylation, but blocks AChR cluster formation. These findings suggest an avenue by which postsynaptic stability can be regulated by modification of intracellular signaling pathways that are distinct from those used during synapse formation.


Subject(s)
Agrin/metabolism , Calcium/metabolism , Egtazic Acid/analogs & derivatives , Receptor Aggregation/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/metabolism , Acetylcholine/pharmacology , Agrin/pharmacology , Animals , Blotting, Western , Bungarotoxins/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Chick Embryo , Dose-Response Relationship, Drug , Drug Interactions , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Fura-2/metabolism , Intracellular Space/metabolism , Muscle Fibers, Skeletal , Phosphorylation , Protein Binding , Rats , Receptors, Cholinergic/drug effects , Synapses/drug effects , Time Factors , Vanadates/pharmacology , Vasodilator Agents/pharmacology
2.
Am J Physiol Cell Physiol ; 280(4): C980-92, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11245615

ABSTRACT

ACh-induced contraction of esophageal circular muscle (ESO) depends on Ca2+ influx and activation of protein kinase Cepsilon (PKCepsilon). PKCepsilon, however, is known to be Ca2+ independent. To determine where Ca2+ is needed in this PKCepsilon-mediated contractile pathway, we examined successive steps in Ca2+-induced contraction of ESO muscle cells permeabilized by saponin. Ca2+ (0.2-1.0 microM) produced a concentration-dependent contraction that was antagonized by antibodies against PKCepsilon (but not by PKCbetaII or PKCgamma antibodies), by a calmodulin inhibitor, by MLCK inhibitors, or by GDPbetas. Addition of 1 microM Ca2+ to permeable cells caused myosin light chain (MLC) phosphorylation, which was inhibited by the PKC inhibitor chelerythrine, by D609 [phosphatidylcholine-specific phospholipase C inhibitor], and by propranolol (phosphatidic acid phosphohydrolase inhibitor). Ca2+-induced contraction and diacylglycerol (DAG) production were reduced by D609 and by propranolol, alone or in combination. In addition, contraction was reduced by AACOCF(3) (cytosolic phospholipase A(2) inhibitor). These data suggest that Ca2+ may directly activate phospholipases, producing DAG and arachidonic acid (AA), and PKCepsilon, which may indirectly cause phosphorylation of MLC. In addition, direct G protein activation by GTPgammaS augmented Ca2+-induced contraction and caused dose-dependent production of DAG, which was antagonized by D609 and propranolol. We conclude that agonist (ACh)-induced contraction may be mediated by activation of phospholipase through two distinct mechanisms (increased intracellular Ca2+ and G protein activation), producing DAG and AA, and activating PKCepsilon-dependent mechanisms to cause contraction.


Subject(s)
Calcium Signaling/physiology , Calcium/pharmacology , Esophagus/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Adrenergic beta-Antagonists/pharmacology , Alkaloids , Animals , Antibodies/pharmacology , Azepines/pharmacology , Benzophenanthridines , Bridged-Ring Compounds/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cats , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Female , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/immunology , Isoenzymes/metabolism , Male , Muscle Contraction/drug effects , Myosin Light Chains/antagonists & inhibitors , Myosin Light Chains/metabolism , Naphthalenes/pharmacology , Norbornanes , Phenanthridines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Phosphorylation , Propranolol/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/immunology , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Pyrrolidinones/pharmacology , Quercetin/pharmacology , Second Messenger Systems/physiology , Thiocarbamates , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
3.
J Physiol ; 531(Pt 2): 315-27, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230506

ABSTRACT

We recorded Ca2+ sparks and spontaneous transient outward currents (STOCs) simultaneously in smooth muscle cells using whole-cell patch recording and a unique, high-speed widefield digital imaging system to monitor fluo-3 fluorescence in both two and three dimensions (2D and 3D). In 2D imaging, the correlation between the amplitude of a spark and its corresponding STOC was a weak one, and 27 % of the sparks failed to cause STOCs. The STOCless sparks were not significantly different in amplitude from those that caused STOCs. Three-dimensional imaging disclosed that STOCless sparks were located close to the cell surface, and on average their apparent distance from the cell surface was not significantly different from the sparks that cause STOCs. Statistical evaluation of spark clusters disclosed that there were regions of the cell where the probability of spark occurrence was high and others where it was quite low.


Subject(s)
Calcium/physiology , Esophagus/physiology , Muscle, Smooth/physiology , Aniline Compounds , Animals , Cats , Cell Membrane/physiology , Electric Conductivity , Esophagus/cytology , Fluorescent Dyes , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Muscle, Smooth/cytology , Patch-Clamp Techniques , Xanthenes
4.
Gastroenterology ; 119(1): 51-61, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10889154

ABSTRACT

BACKGROUND & AIMS: Because tachykinins have been identified as neurotransmitters in the guinea pig colon and human ileum, we examined a possible role of tachykinin receptors and neurokinin (NK) A in neurally induced contraction of human sigmoid colon circular muscle. METHODS: Muscle strips were stimulated electrically for 10 seconds. Single cells were isolated by enzymatic digestion and permeabilized by saponin. [(35)S]GTPgammaS binding was assayed with or without NKA for 5 minutes. Intracellular Ca(2+) was measured using Fura 2. RESULTS: In the presence of 100 micromol/L L-NNA, 100 micromol/L atropine did not affect electrical field stimulation (EFS)-induced contraction. A peptide NK(2)-receptor antagonist (NK-2ra) but not an NK(1) antagonist FK888 (1 micromol/L) eliminated EFS-induced contraction. NKA-induced contraction in muscle strips and single cells was virtually abolished by NK-2ra, but not by FK888. In permeabilized cells, contraction was blocked by Gq-protein antibodies, but not by other G-protein antibodies, suggesting that NKA activates Gq, which was confirmed by a [(35)S]GTPgammaS binding assay. NKA-induced contraction and increase in cytosolic Ca(2+) were abolished by depletion of intracellular Ca(2+) stores. CONCLUSIONS: Tachykinins may be the main excitatory neurotransmitters in human sigmoid circular muscle. NKA activates Gq-linked NK(2) receptors, which cause Ca(2+) release, followed by contraction.


Subject(s)
Colon/physiology , GTP-Binding Proteins/metabolism , Gastrointestinal Motility/physiology , Muscle, Smooth/physiology , Nervous System Physiological Phenomena , Receptors, Neurokinin-2/metabolism , Aged , Aged, 80 and over , Calcium/physiology , Colon/cytology , Colon/innervation , Dipeptides/pharmacology , Electric Stimulation , Female , GTP-Binding Protein alpha Subunits, Gq-G11 , Gastrointestinal Motility/drug effects , Humans , In Vitro Techniques , Indoles/pharmacology , Intracellular Membranes/metabolism , Male , Middle Aged , Muscle, Smooth/innervation , Neurokinin A/pharmacology , Receptors, Neurokinin-2/agonists
5.
J Physiol ; 524 Pt 1: 3-17, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10747180

ABSTRACT

1. A digital imaging microscope with fura-2 as the Ca2+ indicator was used to determine the sources for the rise in intracellular calcium concentration ([Ca2+]i) that occurs when the membrane in a cell-attached patch is stretched. Unitary ionic currents from stretch-activated channels and [Ca2+]i images were recorded simultaneously. 2. When suction was applied to the patch pipette to stretch a patch of membrane, Ca2+-permeable cation channels (stretch-activated channels) opened and a global increase in [Ca2+]i occurred, as well as a greater focal increase in the vicinity of the patch pipette. The global changes in [Ca2+]i occurred only when stretch-activated currents were sufficient to cause membrane depolarization, as indicated by the reduction in amplitude of the unitary currents. 3. When Ca2+ was present only in the pipette solution, just the focal change in [Ca2+]i was obtained. This focal change was not seen when the contribution from Ca2+ stores was eliminated using caffeine and ryanodine. 4. These results suggest that the opening of stretch-activated channels allows ions, including Ca2+, to enter the cell. The entry of positive charge triggers the influx of Ca2+ into the cell by causing membrane depolarization, which presumably activates voltage-gated Ca2+ channels. The entry of Ca2+ through stretch-activated channels is also amplified by Ca2+ release from internal stores. This amplification appears to be greater than that obtained by activation of whole-cell Ca2+ currents. These multiple pathways whereby membrane stretch causes a rise in [Ca2+]i may play a role in stretch-induced contraction, which is a characteristic of many smooth muscle tissues.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Stomach/physiology , Animals , Bufo marinus , In Vitro Techniques , Membrane Potentials , Patch-Clamp Techniques
6.
J Physiol ; 484 ( Pt 2): 331-7, 1995 Apr 15.
Article in English | MEDLINE | ID: mdl-7602529

ABSTRACT

1. using standard single channel patch clamp techniques we studied the stretch sensitivity of a 20 pS K(+)-selective channel which is activated by fatty acids and found in freshly dissociated smooth muscle cells from the stomach of the toad Bufo marinus. 2. A pulse of suction applied to the back of the patch pipette in order to stretch the membrane resulted in activation of this K+ channel. A train of suction pulses resulted in a gradually increased level of channel activity during each successive pulse, as well as an increase in baseline activity between pulses. This pattern contrasts markedly with many other stretch-activated channels whose activation is limited to the duration of the suction pulse. 3. Application of fatty acids augmented the response to stretch. In contrast, application of 10 microM defatted albumin, which removes fatty acids from membranes, rapidly and reversibly decreased the response to stretch. 4. These results are consistent with the hypothesis that fatty acids which are generated by mechanical stimuli, perhaps by mechanically activated phospholipases, are the intermediaries in activation of certain mechanically sensitive ion channels.


Subject(s)
Fatty Acids/pharmacology , Muscle, Smooth/physiology , Potassium Channels/physiology , Albumins/pharmacology , Animals , Bufo marinus , Membrane Potentials/physiology , Myristic Acid , Myristic Acids/pharmacology , Patch-Clamp Techniques , Physical Stimulation , Potassium Channels/drug effects , Time Factors
7.
Article in English | MEDLINE | ID: mdl-7784455

ABSTRACT

A variety of fatty acids increase the activity of certain types of K+ channels. This effect is not dependent on the three enzymatic pathways that convert arachidonic acid to various bioactive oxygenated metabolites. One type of K+ channel in toad stomach smooth muscle cell membranes in activated by fatty acids and other single chain lipids which possess both a negatively charged head group and a sufficiently hydrophobic acyl chain. Neutral lipids have no effect on K+ channel activity, while positively charged lipids with a sufficiently hydrophobic acyl chain suppress channel activity. Acyl Coenzyme A's, which do not flip across the bilayer, act only from the cytosolic surface of the membrane, suggesting that the binding site for channel activation is also located there. This fatty acid-activated channel is also activated by membrane stretch. Moreover, this mechanical response is either mediated or modulated by fatty acids. Thus, fatty acids and other charged single chain lipids may comprise another class of first or second messenger molecules that target ion channels.


Subject(s)
Acyl Coenzyme A/pharmacology , Fatty Acids/pharmacology , Muscle, Smooth/metabolism , Potassium Channels/physiology , Acyl Coenzyme A/chemistry , Animals , Bufo marinus , In Vitro Techniques , Models, Biological , Patch-Clamp Techniques , Physical Stimulation , Stimulation, Chemical , Stomach/cytology
8.
Am J Hypertens ; 7(1): 82-9, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8136116

ABSTRACT

Large-conductance, Ca(2+)-activated K+ channels were identified in single smooth muscle cells freshly isolated from rabbit superior mesenteric artery. They typically showed a reversal potential close to 0 mV in excised, inside-out patches in symmetric 130 mmol/L [K+] with a unitary conductance of 260 pS, and increased activity at more positive potentials and/or when [Ca2+] was raised at the cytosolic surface of the membrane. Both in cell-attached and in excised, inside-out configurations, stretching the membrane patch by applying suction to the back of the patch pipette increased the activity of these channels without changing either the unitary conductance or the voltage sensitivity of the channel. Stretch activation was repeatedly seen in inside-out patches when both surfaces were bathed with a 0 Ca2+ solution containing 2 or 5 mmol/L EGTA to chelate trace amounts of Ca2+, making it highly improbable that stretch activation could be secondary to a stretch-induced flux of Ca2+. Consequently, stretch activation of large-conductance, Ca(2+)-activated K+ channels in mesenteric artery smooth muscle cells seems to be due to a direct effect of stretch on the channel itself or on some closely associated, membrane-bound entity.


Subject(s)
Calcium/physiology , Muscle, Smooth, Vascular/metabolism , Potassium Channels/metabolism , Animals , Cell Membrane/physiology , Cell Membrane Permeability/physiology , Feedback/physiology , Hypertension/physiopathology , In Vitro Techniques , Mesenteric Artery, Superior/cytology , Mesenteric Artery, Superior/metabolism , Muscle Relaxation/physiology , Muscle, Smooth, Vascular/cytology , Rabbits , Second Messenger Systems/physiology , Vascular Resistance/physiology
9.
FEBS Lett ; 297(1-2): 24-8, 1992 Feb 03.
Article in English | MEDLINE | ID: mdl-1551431

ABSTRACT

Large conductance Ca(2+)-activated K+ channels in rabbit pulmonary artery smooth muscle cells are activated by membrane stretch and by arachidonic acid and other fatty acids. Activation by stretch appears to occur by a direct effect of stretch on the channel itself or a closely associated component. In excised inside-out patches stretch activation was seen under conditions which precluded possible mechanisms involving cytosolic factors, release of Ca2+ from intracellular stores, or stretch induced transmembrane flux of Ca2+ or other ions potentially capable of activating the channel. Fatty acids also directly activate this channel. Like stretch activation, fatty acid activation occurs in excised inside-out patches in the absence of cytosolic constituents. Moreover, the channel is activated by fatty acids which, unlike arachidonic acid, are not substrates for the cyclo-oxygenase or lypoxygenase pathways, indicating that oxygenated metabolites do not mediate the response. Thus, four distinct types of stimuli (cytosolic Ca2+, membrane potential, membrane stretch, and fatty acids) can directly affect the activity of this channel.


Subject(s)
Calcium/metabolism , Fatty Acids/pharmacology , Muscle, Smooth, Vascular/metabolism , Potassium Channels/metabolism , Pulmonary Artery/metabolism , Animals , Arachidonic Acid/pharmacology , Cations , Cell Membrane/drug effects , Cell Membrane/physiology , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myristic Acid , Myristic Acids/pharmacology , Potassium Channels/drug effects , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Rabbits
10.
Pflugers Arch ; 417(5): 493-9, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1707158

ABSTRACT

The properties of hyperpolarization-activated channels were studied in single smooth muscle cells from the stomach of the toad, Bufo marinus, using the patch-clamp technique. In cell-attached patches, inward channel currents were activated by hyperpolarizing pulses from a holding potential of -20 mV to potentials more negative than -60 mV. The activity of the channels increased and their latency of activation decreased as the hyperpolarization was increased. The slope conductance of the channels with standard high sodium concentration pipette solution was 64.2 +/- 9.1 pS (SD, n = 17). Stretching the patch, by suction applied to the back of the patch pipette, also increased the activity and shortened the latency of activation. We designate these channels as HA-SACs (hyperpolarization- and stretch-activated channels). HA-SACs were observed in 83% (175/210) of the patches studied. HA-SAC currents were carried by sodium and potassium ions, but their amplitude was increased by replacing extracellular sodium with potassium. Extracellular magnesium and calcium ions significantly reduced the single-channel conductance of HA-SACs. These permeation characteristics and the single-channel conductance of HA-SACs were indistinguishable from those of stretch-activated channels (SACs) previously described in these cells. The following observations are consistent with HA-SACs being a subset of SACs. First, SACs were at times found in cell-attached patches which lacked HA-SACs. Second, the number of channels in a cell-attached patch simultaneously activated by stretch (usually 5-10 and often more) exceeded by far the number simultaneously activated by hyperpolarization (usually one or two).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Ion Channels/physiology , Membrane Potentials/physiology , Muscle, Smooth/physiology , Animals , Bufo marinus , Calcium/physiology , Electric Conductivity , Magnesium/physiology , Muscle, Smooth/metabolism , Potassium/physiology , Sodium/physiology
12.
Pflugers Arch ; 412(4): 339-45, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2459658

ABSTRACT

As in many smooth muscle tissue preparations, single smooth muscle cells freshly dissociated from the stomach of the toad Bufo marinus contract when stretched. Stretch-activated channels have been identified in these cells using patch-clamp techniques. In both cell-attached and excised inside-out patches, the probability of the channel being open (Po) increases when the membrane is stretched by applying negative pressure to the extracellular surface through the patch pipette. The increase in Po is mainly due to a decrease in closed time durations, but an increase in open time duration is also seen. The open-channel current-voltage relationship shows inward rectification and is not appreciably altered when K+ is substituted for Na+ as the charge-carrying cation in Ca2+-free (2 mM EGTA) pipette solutions bathing the extracellular surface of the patch. The inclusion of physiological concentrations of Ca2+ (1.8 mM) in pipette solutions (containing high concentrations of Na+ and low K+) significantly decreases the slope conductance as well as the unitary amplitude. The channel also conducts Ca2+, since inward currents were observed using pipette solutions in which Ca2+ ions were the only inorganic cations. When simulating normal physiological conditions, we find that substantial ionic current is conducted into the cell when the channel is open. These characteristics coupled with the high density of the stretch-activated channels point to a key role for them in the initiation of stretch-induced contraction.


Subject(s)
Ion Channels/physiology , Muscle Contraction , Muscle, Smooth/physiology , Animals , Bufo marinus , In Vitro Techniques , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Stomach/cytology
13.
J Theor Biol ; 116(1): 111-26, 1985 Sep 07.
Article in English | MEDLINE | ID: mdl-2413315

ABSTRACT

Certain macromolecules embedded in the cell membranes of a variety of cells behave as gated ion-selective pores or channels. The length of time that a channel remains open or closed is not deterministic in nature and must be described in terms of relative probabilities. If channels act independently of each other and appropriate experimental conditions can be maintained, the behavior of a channel can be described by a homogeneous Markov process. Using this representation, the relative probability of observing openings (or closings) of various durations can be described by a sum of discrete components which are related to the underlying model of the kinetic behavior of the channel. Generally, these discrete components are taken to be simple decaying exponentials; however, exponentially decaying oscillatory components (as well as certain others which are discussed) are consistent with the Markov process representation. The presence of components other than simple decaying exponentials is shown to imply the violation of detailed balance in the steady-state (which requires energy), and thus, the presence of cyclic pathways in models which accurately represent the kinetic behavior of the channel. Oscillatory components, if present, will in general decay at a faster rate than the slowest decaying component, which, except under a very restricted set of conditions, will be a simple exponential.


Subject(s)
Ion Channels , Cell Membrane Permeability , Markov Chains , Mathematics , Probability
SELECTION OF CITATIONS
SEARCH DETAIL