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










Publication year range
1.
J Neurophysiol ; 115(1): 345-54, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26561606

ABSTRACT

Chronic intermittent hypoxia (CIH) is a hallmark manifestation of sleep apnea. A heightened carotid body activity and the resulting chemosensory reflex mediate increased sympathetic nerve activity by CIH. However, the mechanisms underlying heightened carotid body activity by CIH are not known. An elevation of intracellular calcium ion concentration ([Ca(2+)]i) in glomus cells, the primary oxygen-sensing cells, is an essential step for carotid body activation by hypoxia. In the present study, we examined the effects of CIH on the glomus cell [Ca(2+)]i response to hypoxia and assessed the underlying mechanisms. Glomus cells were harvested from adult rats or wild-type mice treated with 10 days of either room air (control) or CIH (alternating cycles of 15 s of hypoxia and 5 min of room air; 9 episodes/h; 8 h/day). CIH-treated glomus cells exhibited an enhanced [Ca(2+)]i response to hypoxia, and this effect was absent in the presence of 2-(4-cyclopropylphenyl)-N-((1R)-1-[5-[(2,2,2-trifluoroethyl)oxo]-pyridin-2-yl]ethyl)acetamide (TTA-A2), a specific inhibitor of T-type Ca(2+) channels, and in voltage-gated calcium channel, type 3.2 (CaV3.2), null glomus cells. CaV3.2 knockout mice exhibited an absence of CIH-induced hypersensitivity of the carotid body. CIH increased reactive oxygen species (ROS) levels in glomus cells. A ROS scavenger prevented the exaggerated TTA-A2-sensitive [Ca(2+)]i response to hypoxia. CIH had no effect on CaV3.2 mRNA levels. CIH augmented Ca(2+) currents and increased CaV3.2 protein in plasma membrane fractions of human embryonic kidney-293 cells stably expressing CaV3.2, and either a ROS scavenger or brefeldin-A, an inhibitor of protein trafficking, prevented these effects. These findings suggest that CIH leads to an augmented Ca(2+) influx via ROS-dependent facilitation of CaV3.2 protein trafficking to the plasma membrane.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium/metabolism , Carotid Body/metabolism , Hypoxia/metabolism , Animals , Benzeneacetamides/administration & dosage , Calcium Channels, T-Type/physiology , Carotid Body/drug effects , Cell Hypoxia , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Transport , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
2.
Adv Exp Med Biol ; 661: 155-67, 2010.
Article in English | MEDLINE | ID: mdl-20204729

ABSTRACT

Oxidative [Au1]stress, through the production of oxygen metabolites such as hydrogen peroxide[Au2] (H(2)O(2)), increases vascular endothelial permeability and plays a crucial role in several lung diseases. The transient receptor potential (melastatin) 2 (TRPM2) is an oxidant-sensitive, nonselective cation channel that is widely expressed in mammalian tissues, including the vascular endothelium. We have demonstrated the involvement of TRPM2 in mediating oxidant-induced calcium entry and endothelial hyperpermeability in cultured pulmonary artery endothelial cells. Here, we provide evidence that neutrophil activation-dependent increase in endothelial permeability and neutrophil extravasation requires TRPM2 in cultured endothelial cells. In addition, protein kinase Calpha (PKCalpha) that rapidly colocalizes with the short (nonconducting) TRPM2 isoform after exposure to hydrogen peroxide positively regulates calcium entry through the functional TRPM2 channel. Thus, increase in lung microvessel permeability and neutrophil sequestration depends on the activation of endothelial TRPM2 by neutrophilic oxidants and on PKCalpha regulation of TRPM2 channel activity. Manipulating TRPM2 function in the endothelium may represent a novel strategy aimed to prevent oxidative stress-related vascular dysfunction.


Subject(s)
Endothelial Cells/metabolism , TRPM Cation Channels/metabolism , Animals , Calcium/metabolism , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/cytology , Hydrogen Peroxide/metabolism , Inflammation/metabolism , Lung/blood supply , Lung/metabolism , Lung/pathology , Neutrophils/immunology , Oxidants/metabolism , Oxidative Stress , Protein Kinase C-alpha/metabolism , TRPM Cation Channels/genetics
3.
Circ Res ; 102(3): 347-55, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18048770

ABSTRACT

Oxidative stress through the production of oxygen metabolites such as hydrogen peroxide (H2O2) increases vascular endothelial permeability. H2O2 stimulates ADP-ribose formation, which in turn opens transient receptor potential melastatin (TRPM)2 channels. Here, in endothelial cells, we demonstrate transcript and protein expression of TRPM2, a Ca2+-permeable, nonselective cation channel. We further show the importance of TRPM2 expression in signaling of increased endothelial permeability by oxidative stress. Exposure of endothelial cell monolayers to sublytic concentrations of H2O2 induced a cationic current measured by patch-clamp recording and Ca2+ entry detected by intracellular fura-2 fluorescence. H2O2 in a concentration-dependent manner also decreased trans-monolayer transendothelial electrical resistance for 3 hours (with maximal effect seen at 300 micromol/L H2O2), indicating opening of interendothelial junctions. The cationic current, Ca2+ entry, and transendothelial electrical resistance decrease elicited by H2O2 were inhibited by siRNA depleting TRPM2 or antibody blocking of TRPM2. H2O2 responses were attenuated by overexpression of the dominant-negative splice variant of TRPM2 or inhibition of ADP-ribose formation. Overexpression of the full-length TRPM2 enhanced H2O2-mediated Ca2+ entry, cationic current, and the transendothelial electrical resistance decrease. Thus, TRPM2 mediates H2O2-induced increase in endothelial permeability through the activation of Ca2+ entry via TRPM2. TRPM2 represents a novel therapeutic target directed against oxidant-induced endothelial barrier disruption.


Subject(s)
Calcium/metabolism , Capillary Permeability/drug effects , Endothelial Cells/metabolism , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , TRPM Cation Channels/metabolism , Adenosine Diphosphate Ribose/antagonists & inhibitors , Adenosine Diphosphate Ribose/metabolism , Capillary Permeability/physiology , Cations, Divalent/metabolism , Cells, Cultured , Electric Impedance , Endothelial Cells/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Hydrogen Peroxide/metabolism , Ion Transport/drug effects , Ion Transport/physiology , Oxidants/metabolism , Patch-Clamp Techniques , RNA, Small Interfering/pharmacology , TRPM Cation Channels/antagonists & inhibitors
4.
J Biol Chem ; 282(11): 7833-43, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17197445

ABSTRACT

RhoA activation and increased intracellular Ca(2+) concentration mediated by the activation of transient receptor potential channels (TRPC) both contribute to the thrombin-induced increase in endothelial cell contraction, cell shape change, and consequently to the mechanism of increased endothelial permeability. Herein, we addressed the possibility that TRPC signals RhoA activation and thereby contributes in actinomyosin-mediated endothelial cell contraction and increased endothelial permeability. Transduction of a constitutively active Galphaq mutant in human pulmonary arterial endothelial cells induced RhoA activity. Preventing the increase in intracellular Ca2+ concentration by the inhibitor of Galphaq or phospholipase C and the Ca2+ chelator, BAPTA-AM, abrogated thrombin-induced RhoA activation. Depletion of extracellular Ca2+ also inhibited RhoA activation, indicating the requirement of Ca2+ entry in the response. RhoA activation could not be ascribed to storeoperated Ca2+ (SOC) entry because SOC entry induced with thapsigargin or small interfering RNA-mediated inhibition of TRPC1 expression, the predominant SOC channel in these endothelial cells, failed to alter RhoA activity. However, activation of receptor-operated Ca2+ entry by oleoyl-2-acetyl-sn-glycerol, the membrane permeable analogue of the Galphaq-phospholipase C product diacylglycerol, induced RhoA activity. Receptor-operated Ca2+ activation was mediated by TRPC6 because small interfering RNA-induced TRPC6 knockdown significantly reduced Ca2+ entry. TRPC6 knockdown also prevented RhoA activation, myosin light chain phosphorylation, and actin stress fiber formation as well as inter-endothelial junctional gap formation in response to either oleoyl-2-acetyl-sn-glycerol or thrombin. TRPC6-mediated RhoA activity was shown to be dependent on PKCalpha activation. Our results demonstrate that Galphaq activation of TRPC6 signals the activation of PKCalpha, and thereby induces RhoA activity and endothelial cell contraction.


Subject(s)
Calcium/metabolism , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , TRPC Cation Channels/metabolism , Thrombin/metabolism , rhoA GTP-Binding Protein/metabolism , Biological Transport , Cells, Cultured , Chelating Agents/pharmacology , Endothelium, Vascular/cytology , Humans , Protein Kinase C-alpha/metabolism , Pulmonary Artery/cytology , Signal Transduction , TRPC6 Cation Channel , Type C Phospholipases/metabolism
5.
Microcirculation ; 13(8): 693-708, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17085428

ABSTRACT

Increased endothelial permeability is the hallmark of inflammatory vascular edema. Inflammatory mediators that bind to heptahelical G protein-coupled receptors trigger increased endothelial permeability by increasing the intracellular Ca2+ concentration ([Ca2+]i). The rise in [Ca2+]i activates key signaling pathways that mediate cytoskeletal reorganization (through myosin-light-chain-dependent contraction) and the disassembly of VE-cadherin at the adherens junctions. The Ca2+-dependent protein kinase C (PKC) isoform PKCalpha plays a crucial role in initiating endothelial cell contraction and disassembly of VE-cadherin junctions. The increase in [Ca2+]i induced by inflammatory agonists such as thrombin and histamine is achieved by the generation of inositol 1,4,5-trisphosphate (IP3), activation of IP3-receptors, release of stored intracellular Ca2+, and Ca2+ entry through plasma membrane channels. IP3-sensitive Ca2+-store depletion activates plasma membrane cation channels (i.e., store-operated cation channels [SOCs] or Ca2+ release-activated channels [CRACs]) to cause Ca2+ influx into endothelial cells. Recent studies have identified members of Drosophila transient receptor potential (TRP) gene family of channels that encode functional SOCs in endothelial cells. These studies also suggest that the canonical TRPC homologue TRPC1 is the predominant isoform expressed in human vascular endothelial cells, and is the essential component of the SOC in this cell type. Further, evidence suggests that the inflammatory cytokine tumor necrosis factor-alpha can induce the expression of TRPC1 in human vascular endothelial cells signaling via the nuclear factor-kappaB pathway. Increased expression of TRPC1 augments Ca2+ influx via SOCs and potentiates the thrombin-induced increase in permeability in human vascular endothelial cells. Deletion of the canonical TRPC homologue in mouse, TRPC4, caused impairment in store-operated Ca2+ current and Ca2+-store release-activated Ca2+ influx in aortic and lung endothelial cells. In TRPC4 knockout (TRPC4-/-) mice, acetylcholine-induced endothelium-dependent smooth muscle relaxation was drastically reduced. In addition, TRPC4-/- mouse-lung endothelial cells exhibited lack of actin-stress fiber formation and cell retraction in response to thrombin activation of protease-activated receptor-1 (PAR-1) in endothelial cells. The increase in lung microvascular permeability in response to PAR-1 activation was inhibited in TRPC4-/- mice. These results indicate that endothelial TRP channels such as TRPC1 and TRPC4 play an important role in signaling agonist-induced increases in endothelial permeability.


Subject(s)
Calcium Signaling , Calcium/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Edema/metabolism , Humans , Inflammation/metabolism , Mice , Permeability
6.
Pflugers Arch ; 451(1): 131-42, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15988589

ABSTRACT

The endothelial cells (ECs) form a semipermeable barrier between the blood and the tissue. An important function of the endothelium is to maintain the integrity of the barrier function of the vessel wall. Ca(2+) signaling in ECs plays a key role in maintaining the barrier integrity. Transient receptor potential canonical (TRPC) channels are mammalian homologs of Drosophila TRP Ca(2+)-permeable channels expressed in EC. TRPC channels are thought to function as a Ca(2+) entry channel operated by store-depletion as well as receptor-activated channels in a variety of cell types, including ECs. Inflammatory mediators such as thrombin, histamine, bradykinin, and others increase endothelial permeability by actin polymerization-dependent EC rounding and formation of inter-endothelial gaps, a process critically dependent on the increase in EC cytosolic [Ca(2+)] ([Ca(2+)](i)). Increase in endothelial permeability depends on both intracellular Ca(2+) release and extracellular Ca(2+) entry through TRPC channels. This review summarizes recent findings on the role of TRPC channels in the mechanism of Ca(2+) entry in ECs, and, in particular, the role of TRPC channels in regulating endothelial barrier function.


Subject(s)
Calcium Signaling/physiology , Cell Membrane Permeability/physiology , Endothelial Cells/physiology , TRPC Cation Channels/physiology , Animals , Calcium/metabolism , Calcium Channels/physiology , Humans , Inositol 1,4,5-Trisphosphate Receptors , Phosphorylation , Protein Interaction Mapping , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , TRPC6 Cation Channel
7.
J Biol Chem ; 280(17): 17320-8, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15728185

ABSTRACT

Sphingosine 1-phosphate (S1P) ligation of endothelial differentiation gene-1 receptor coupled to the heterotrimeric G protein, Gi, promotes endothelial barrier strengthening via Rac-dependent assembly of adherens junctions (AJs). However, the mechanism of Rac activation induced by S1P stimulation remains unclear. In live endothelial cells expressing GFP-Rac, we observed that S1P induced the translocation of Rac to intercellular junctions, resulting in junctional sealing. We investigated the role of intracellular Ca2+ in signaling Rac activation and the enhancement of endothelial barrier function. We observed that S1P activated the release of Ca2+ from endoplasmic reticulum stores, and subsequent Ca2+ entry via lanthanum-sensitive store-operated Ca2+ channels (SOC) after store depletion. Inhibition of Gi, phospholipase C, or inositol trisphosphate receptor prevented the S1P-activated increase in intracellular Ca2+ as well as Rac activation, AJ assembly, and enhancement of endothelial barrier. Chelation of intracellular Ca2+ with BAPTA blocked S1P-induced Rac activation, indicating the requirement for Ca2+ in the response. Inhibition of SOC by lanthanum or transient receptor potential channel 1 (TRPC1), a SOC constituent, by TRPC1 antibody, failed to prevent S1P-induced Rac translocation to junctions and AJ assembly. Thus, our results demonstrate that S1P promotes endothelial junctional integrity by activating the release of endoplasmic reticulum-Ca2+, which induces Rac activation and promotes AJ annealing.


Subject(s)
Adherens Junctions/metabolism , Egtazic Acid/analogs & derivatives , Endothelial Cells/cytology , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Animals , Aorta/metabolism , Blotting, Western , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Coloring Agents/pharmacology , Egtazic Acid/pharmacology , Electric Impedance , Electrophysiology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Enzyme Activation , Gap Junctions , Humans , Inositol 1,4,5-Trisphosphate Receptors , Microscopy, Fluorescence , Patch-Clamp Techniques , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Lysosphingolipid/metabolism , Time Factors , Transfection , Type C Phospholipases/metabolism , rac GTP-Binding Proteins/metabolism
8.
J Biol Chem ; 280(5): 3178-84, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15576373

ABSTRACT

Endothelial cells exhibit regulated exocytosis in response to inflammatory mediators such as thrombin and histamine. The exocytosis of Weibel-Palade bodies (WPBs) containing von Willebrand factor, P-selectin, and interleukin-8 within minutes after stimulation is important for vascular homeostasis. SNARE proteins are key components of the exocytic machinery in neurons and some secretory cells, but their role in regulating exocytosis in endothelial cells is not well understood. We examined the function of SNARE proteins in mediating exocytosis of WPBs in endothelial cells. We identified the presence of syntaxin 4, syntaxin 3, and the high affinity syntaxin 4-regulatory protein Munc18c in human lung microvascular endothelial cells. Small interfering RNA-induced knockdown of syntaxin 4 (but not of syntaxin 3) inhibited exocytosis of WPBs as detected by the reduction in thrombin-induced cell surface P-selectin expression. Thrombin ligation of protease-activated receptor-1 activated the phosphorylation of syntaxin 4 and Munc18c, which, in turn, disrupted the interaction between syntaxin 4 and Munc18. Protein kinase Calpha activation was required for the phosphorylation of syntaxin 4 and Munc18c as well as the cell surface expression of P-selectin. We also observed that syntaxin 4 knockdown inhibited the adhesion of neutrophils to thrombin-activated endothelial cells, demonstrating the functional role of syntaxin 4 in promoting endothelial adhesivity. Thus, protease-activated receptor-1-induced protein kinase Calpha activation and phosphorylation of syntaxin 4 and Munc18c are required for the cell surface expression of P-selectin and the consequent binding of neutrophils to endothelial cells.


Subject(s)
Endothelium, Vascular/enzymology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Kinase C/metabolism , Receptor, PAR-1/metabolism , Vesicular Transport Proteins/metabolism , Animals , Calcium/metabolism , Cell Adhesion/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Exocytosis/physiology , Hemostatics/pharmacology , Humans , Lung/blood supply , Microcirculation , Munc18 Proteins , Neutrophils/cytology , P-Selectin/metabolism , Phosphorylation , Protein Kinase C-alpha , Qa-SNARE Proteins , Rabbits , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombin/pharmacology
9.
Am J Physiol Lung Cell Mol Physiol ; 287(6): L1303-13, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15347566

ABSTRACT

We determined the effects of TNF-alpha on the expression of transient receptor potential channel (TRPC) homologues in human vascular endothelial cells and the consequences of TRPC expression on the endothelial permeability response. We observed that TNF-alpha exposure increased TRPC1 expression without significantly altering expression of other TRPC isoforms in human pulmonary artery endothelial cells (HPAEC). Because TRPC1 belongs to the store-operated cation channel family, we measured the Ca(2+) store depletion-mediated Ca(2+) influx in response to thrombin exposure. We observed that thrombin-induced Ca(2+) influx in TNF-alpha-stimulated HPAEC was twofold greater than in control cells. To address the relationship between store-operated Ca(2+) influx and TRPC1 expression, we overexpressed TRPC1 by three- to fourfold in the human dermal microvascular endothelial cell line (HMEC) using the TRPC1 cDNA. Thrombin-induced store Ca(2+) depletion in these cells caused approximately twofold greater increase in Ca(2+) influx than in control cells. Furthermore, the inositol 1,4,5-trisphosphate-sensitive store-operated cationic current was increased greater than twofold in TRPC1-transfected cells compared with control. To address the role of Ca(2+) influx via TRPC1 in signaling endothelial permeability, we measured actin-stress fiber formation and transendothelial monolayer electrical resistance (TER) in the TRPC1 cDNA-transfected HMEC and TNF-alpha-challenged HPAEC. Both thrombin-induced actin-stress fiber formation and a decrease in TER were augmented in TRPC1-overexpressing HMEC compared with control cells. TNF-alpha-induced increased TRPC1 expression in HPAEC also resulted in marked endothelial barrier dysfunction in response to thrombin. These findings indicate the expression level of TRPC1 in endothelial cells is a critical determinant of Ca(2+) influx and signaling of the increase in endothelial permeability.


Subject(s)
Calcium Channels/genetics , Calcium/physiology , Cell Membrane Permeability/physiology , Endothelium, Vascular/physiology , Gene Expression Regulation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Sequence , Calcium Channels/drug effects , Calcium Channels/metabolism , Cell Membrane Permeability/drug effects , DNA Primers , DNA, Complementary/genetics , Endothelium, Vascular/drug effects , Humans , Pulmonary Artery , Rabbits , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels , Transfection
10.
Am J Physiol Cell Physiol ; 287(2): C527-38, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15084477

ABSTRACT

Palytoxin is a coral toxin that seriously impairs heart function, but its effects on excitation-contraction (E-C) coupling have remained elusive. Therefore, we studied the effects of palytoxin on mechanisms involved in atrial E-C coupling. In field-stimulated cat atrial myocytes, palytoxin caused elevation of diastolic intracellular Ca(2+) concentration ([Ca(2+)](i)), a decrease in [Ca(2+)](i) transient amplitude, Ca(2+) alternans followed by [Ca(2+)](i) waves, and failures of Ca(2+) release. The decrease in [Ca(2+)](i) transient amplitude occurred despite high sarcoplasmic reticulum (SR) Ca(2+) load. In voltage-clamped myocytes, palytoxin induced a current with a linear current-voltage relationship (reversal potential approximately 5 mV) that was blocked by ouabain. Whole cell Ca(2+) current and ryanodine receptor Ca(2+) release channel function remained unaffected by the toxin. However, palytoxin significantly reduced Ca(2+) pumping of isolated SR vesicles. In current-clamped myocytes stimulated at 1 Hz, palytoxin induced a depolarization of the resting membrane potential that was accompanied by delayed afterdepolarizations. No major changes of action potential configuration were observed. The results demonstrate that palytoxin interferes with the function of the sarcolemmal Na(+)-K(+) pump and the SR Ca(2+) pump. The suggested mode of palytoxin toxicity in the atrium involves the conversion of Na(+)-K(+) pumps into nonselective cation channels as a primary event followed by depolarization, Na(+) accumulation, and Ca(2+) overload, which, in turn, causes arrhythmogenic [Ca(2+)](i) waves and delayed afterdepolarizations.


Subject(s)
Acrylamides/pharmacology , Calcium-Transporting ATPases/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cats , Cnidarian Venoms , Electric Stimulation , Heart Atria/cytology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Ouabain/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases
11.
J Biol Chem ; 279(20): 20941-9, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15016832

ABSTRACT

The TRPC1 (transient receptor potential canonical-1) channel is a constituent of the nonselective cation channel that mediates Ca2+ entry through store-operated channels (SOCs) in human endothelial cells. We investigated the role of protein kinase Calpha (PKCalpha) phosphorylation of TRPC1 in regulating the opening of SOCs. Thrombin or thapsigargin added to the external medium activated Ca2+ entry after Ca2+ store depletion, which we monitored by changes in cellular Fura 2 fluorescence. Internal application of the metabolism-resistant analog of inositol 1,4,5-trisphosphate (IP3) activated an inward cationic current within 1 min, which we recorded using the whole cell patch clamp technique. La3+ or Gd3+ abolished the current, consistent with the known properties of SOCs. Pharmacological (Gö6976) or genetic (kinase-defective mutant) inhibition of PKCalpha markedly inhibited IP3-induced activation of the current. Thrombin or thapsigargin also activated La3+-sensitive Ca2+ entry in a PKCalpha-dependent manner. We determined the effects of a specific antibody directed against an extracellular epitope of TRPC1 to address the functional importance of TRPC1. External application of the antibody blocked thrombin- or IP3-induced Ca2+ entry. In addition, we showed that addithrombin or thapsigargin induced phosphorylation of TRPC1 within 1 min. Thrombin failed to induce TRPC1 phosphorylation in the absence of PKCalpha activation. Phosphorylation of TRPC1 and the resulting Ca2+ entry were essential for the increase in permeability induced by thrombin in confluent endothelial monolayers. These results demonstrate that PKCalpha phosphorylation of TRPC1 is an important determinant of Ca2+ entry in human endothelial cells.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Endothelium, Vascular/physiology , Protein Kinase C/metabolism , Antibodies/pharmacology , Biological Transport , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Kinetics , Phosphorylation , Protein Kinase C-alpha , TRPC Cation Channels , Thapsigargin/pharmacology , Umbilical Veins
12.
J Biol Chem ; 278(35): 33492-500, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12766172

ABSTRACT

We tested the hypothesis that RhoA, a monomeric GTP-binding protein, induces association of inositol trisphosphate receptor (IP3R) with transient receptor potential channel (TRPC1), and thereby activates store depletion-induced Ca2+ entry in endothelial cells. We showed that RhoA upon activation with thrombin associated with both IP3R and TRPC1. Thrombin also induced translocation of a complex consisting of Rho, IP3R, and TRPC1 to the plasma membrane. IP3R and TRPC1 translocation and association required Rho activation because the response was not seen in C3 transferase (C3)-treated cells. Rho function inhibition using Rho dominant-negative mutant or C3 dampened Ca2+ entry regardless of whether Ca2+ stores were emptied by thrombin, thapsigargin, or inositol trisphosphate. Rho-induced association of IP3R with TRPC1 was dependent on actin filament polymerization because latrunculin (which inhibits actin polymerization) prevented both the association and Ca2+ entry. We also showed that thrombin produced a sustained Rho-dependent increase in cytosolic Ca2+ concentration [Ca2+]i in endothelial cells overexpressing TRPC1. We further showed that Rho-activated Ca2+ entry via TRPC1 is important in the mechanism of the thrombin-induced increase in endothelial permeability. In summary, Rho activation signals interaction of IP3R with TRPC1 at the plasma membrane of endothelial cells, and triggers Ca2+ entry following store depletion and the resultant increase in endothelial permeability.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/physiology , Calcium/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/pharmacology , Actins/chemistry , Botulinum Toxins/pharmacology , Calcium Channels/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genes, Dominant , Humans , Inositol 1,4,5-Trisphosphate Receptors , Microscopy, Confocal , Models, Biological , Patch-Clamp Techniques , Precipitin Tests , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , TRPC Cation Channels , Thapsigargin/chemistry , Thapsigargin/pharmacology , Thrombin/chemistry , Time Factors , Transfection , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/chemistry
13.
Vascul Pharmacol ; 38(3): 131-41, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12402511

ABSTRACT

The effects of moricizine on Na+ channel currents (INa) were investigated in guinea-pig atrial myocytes and its effects on INa in ventricular myocytes and on cloned hH1 current were compared using the whole-cell, patch-clamp technique. Moricizine induced the tonic block of INa with the apparent dissociation constant (Kd,app) of 6.3 microM at -100 mV and 99.3 microM at -140 mV. Moricizine at 30 microM shifted the h infinity curve to the hyperpolarizing direction by 8.6 +/- 2.4 mV. Moricizine also produced the phasic block of INa, which was enhanced with the increase in the duration of train pulses, and was more prominent with a holding potential (HP) of -100 mV than with an HP of -140 mV. The onset block of INa induced by moricizine during depolarization to -20 mV was continuously increased with increasing the pulse duration, and was enhanced at the less negative HP. The slower component of recovery of the moricizine-induced INa block was relatively slow, with a time constant of 4.2 +/- 2.0 s at -100 mV and 3.0 +/- 1.2 s at -140 mV. Since moricizine induced the tonic block of ventricular INa with Kd,app of 3.1 +/- 0.8 microM at HP = -100 mV and 30.2 +/- 6.8 microM at HP = -140 mV, and cloned hH1 with Kd,app of 3.0 +/- 0.5 microM at HP = -100 mV and 22.0 +/- 3.2 microM at HP = -140 mV, respectively, either ventricular INa or cloned hH1 had significantly higher sensitivity to moricizine than atrial INa. The h infinity curve of ventricular INa was shifted by 10.5 +/- 3.5 mV by 3 microM moricizine and that of hH1 was shifted by 5.0 +/- 2.3 mV by 30 microM moricizine. From the modulated receptor theory, we have estimated the dissociation constants for the resting and inactivated state to be 99.3 and 1.2 microM in atrial myocytes, 30 and 0.17 microM in ventricular myocytes, and 22 and 0.2 microM in cloned hH1, respectively. We conclude that moricizine has a higher affinity for the inactivated Na+ channel than for the resting state channel in atrial myocytes, and moricizine showed the significant atrioventricular difference of moricizine block on INa. Moricizine would exert an antiarrhythmic action on atrial myocytes, as well as on ventricular myocytes, by blocking Na+ channels with a high affinity to the inactivated state and a slow dissociation kinetics.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Moricizine/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Animals , Cells, Cultured , Guinea Pigs , Heart Atria/cytology , Heart Ventricles/cytology , Kinetics , Models, Cardiovascular , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Sodium Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...