Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Urol Int ; 86(2): 220-7, 2011.
Article in English | MEDLINE | ID: mdl-21051874

ABSTRACT

INTRODUCTION: Migration of cells involves a complex signaling network. The aim of the present study was to elucidate the impact of Rho-kinase (ROK) on G protein-coupled receptor-induced migration of human transitional cell carcinoma cells in an in vitro experimental setting. MATERIALS AND METHODS: Intracellular calcium concentration ([Ca(2+)](i)) was measured with the indicator dye Fura-2 in response to lysophosphatidic acid, thrombin and sphingosine-1-phosphate. Phospholipase C activity was determined in myo-[(3)H]inositol- (0.5 µCi/ml) labeled cells. Migration was performed using a Boyden chamber. Transient transfection of a dominant-negative mutant of ROK was done with calcium phosphate. For staining of actin filaments, tetramethylrhodamine isothiocyanate-conjugated phalloidin was used. RESULTS: Lysophosphatidic acid, thrombin and sphingosine-1-phosphate cause increases in [Ca(2+)](i), cellular responses being accompanied by an enhancement of phospholipase C activity and sensitive to the G(i) inhibitor pertussis toxin. Agonists potently stimulated migration of T24 and J82 cells. Inhibition of Rho proteins by Clostridium difficile toxin B abrogated cell migration. Inhibition of ROK using HA1077 and Y-27632 mimicked the properties of toxin B. Expression of a ROK mutant drastically reduced migration. CONCLUSIONS: G protein-coupled receptors potently stimulated cell migration in T24 and J82 cells. Rho proteins and ROK play a pivotal role in this signaling cascade. Rho and ROK may be putative targets for new therapy options in bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/enzymology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Actins/chemistry , Amides/pharmacology , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Calcium/chemistry , Calcium/metabolism , Cell Line, Tumor , Cell Movement , Fura-2/pharmacology , Gene Expression Regulation, Neoplastic , Humans , In Vitro Techniques , Lysophospholipids/chemistry , Lysophospholipids/pharmacology , Mutation , Pertussis Toxin/pharmacology , Phalloidine/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Type C Phospholipases/metabolism , rho-Associated Kinases/metabolism
2.
Thromb Haemost ; 103(3): 662-78, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20135060

ABSTRACT

Membrane-permeable 8-(4-chlorophenylthio)-2'-O-methyl cyclic AMP (8-pCPT-2'-O-Me-cAMP) has been shown to specifically activate cAMP-regulated Epac proteins, without direct effects on protein kinase A and protein kinase G. During isometric tension measurements in thoracic aortic rings from Wistar rats, we observed that 8-pCPT-2'-O-Me-cAMP selectively induced a rightward shift of the concentration response curve for the thromboxane mimetic U46619, without altering the contractile response to noradrenaline. We hypothesised that 8-pCPT-2'-O-Me-cAMP and similar compounds may function as direct thromboxane receptor antagonists. Indeed, in addition to 8-pCPT-2'-O- Me-cAMP, also 8-pCPT-cAMP, 8-(4-chlorophenylthio)-adenosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-8-CPT-cAMPS) and 8-CPT-adenosine, but not 8-Bromo-2'-O-Me-cAMP, induced rightward shifts of the contractile response to U46619. Likewise, 8-pCPT-2'-O- Me-cAMP and Rp-8-CPT-cAMPS, but not 8-Bromo-2'-O-Me-cAMP, specifically reduced U46619-induced aggregation of human platelets. In addition, 8-pCPT-2'-O-Me-cAMP and Rp-8-CPT-cAMPS completely reversed U46619-induced reduction of intercellular adhesion molecule-1 expression and migration of human coronary artery endothelial cells. Most important, the cAMP analogs that reduced the contractile response to U46619 also concentration-dependently inhibited binding of the thromboxane receptor radioligand [5,6-3H]SQ29548 to human platelets. We conclude that 8-pCPT-conjugated cAMP analogs exert competitive thromboxane receptor antagonistic properties.


Subject(s)
Cyclic AMP/analogs & derivatives , Receptors, Thromboxane/antagonists & inhibitors , Animals , Aorta/physiology , Cell Adhesion Molecules , Cell Movement/drug effects , Coronary Vessels/cytology , Cyclic AMP/pharmacology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Platelet Aggregation/drug effects , Rats , Rats, Wistar , Vasoconstriction/drug effects
3.
Cell Signal ; 22(3): 476-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19913094

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates cell growth and survival, migration and adhesion in many cell types. S1P is generated by sphingosine kinases (SphKs), and dephosphorylated by phosphatases or cleaved by S1P lyase. Extracellular S1P activates specific G protein-coupled receptors while intracellular S1P can mobilize Ca2+ from thapsigargin-sensitive stores. Here, we have studied Ca2+ signalling in mouse embryonic fibroblasts (MEFs) deficient in S1P lyase. In these cells, S1P and sphingosine concentrations were elevated about 6-fold and 2-fold, respectively, as measured by liquid chromatography/tandem mass spectrometry. Measurements with fura-2-loaded cells in suspension revealed that resting [Ca2+]i was elevated and agonist-induced [Ca2+]i increases were augmented in S1P lyase-deficient MEFs both in the presence and absence of extracellular Ca2+. Importantly, [Ca2+]i increases and Ca2+ mobilization induced by the SERCA inhibitor, thapsigargin, were augmented, indicating enhanced Ca2+ storage in S1P lyase-deficient MEFs. Measurements with single cells expressing the calmodulin-based Ca2+ sensor, cameleon, revealed that at least two cell types could be distinguished in both MEF cell populations, one with a rapid and transient [Ca2+]i increase and the other with a slower and prolonged [Ca2+]i elevation upon stimulation with thapsigargin. The area under the time course of thapsigargin-induced [Ca2+]i increases, reflecting overall Ca2+ release, was significantly increased by more than 50% in both rapidly and slowly responding S1P lyase-deficient cells. It is concluded that elevated concentrations of S1P and/or sphingosine lead to enhanced Ca2+ storage and elevated basal [Ca2+]i. S1P metabolism thus plays a role not only in acute Ca2+ mobilization but also in long-term regulation of Ca2+ homeostasis.


Subject(s)
Aldehyde-Lyases/metabolism , Calcium/metabolism , Fibroblasts/metabolism , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Animals , Calcium Signaling , Calmodulin/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Lysophospholipids/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thapsigargin/pharmacology
4.
Biochim Biophys Acta ; 1791(5): 357-70, 2009 May.
Article in English | MEDLINE | ID: mdl-19830907

ABSTRACT

Sphingosine-1-phosphate (S1P), formed by sphingosine kinases (SphKs), regulates cellular proliferation and migration by acting as an agonist at specific receptors or intracellularly. Since S1P's effects are probably dependent on subcellular localization of its formation and degradation, we have studied the influence of G protein-coupled receptors on the localization of SphK1. Activation of Gq-coupled receptors induced a profound, rapid (half-life 3-5 s) and long-lasting (> 2 h) translocation of SphK1 to the plasma membrane. This was mimicked by expression of constitutively active G protein alpha-subunits specifically of the Gq family. Classical Gq signalling pathways, or phosphorylation at Ser225, phospholipase D and Ca2+/calmodulin were not involved in M3 receptor-induced SphK1 translocation in HEK-293 cells. Translocation was associated with S1P receptor internalization, which was dependent on catalytic activity of SphK1 and S1P receptor binding and thus resulted from S1P receptor cross-activation. It is concluded that SphK1 is an important effector of Gq-coupled receptors, linking them via cross-activation of S1P receptors to G(i) and G12/13 signalling pathways.


Subject(s)
Cell Membrane/enzymology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Cell Membrane/drug effects , Diglycerides/metabolism , Endocytosis/drug effects , Humans , Mice , Phospholipase D/metabolism , Phosphoserine/metabolism , Protein Kinase C/metabolism , Protein Transport/drug effects , Receptor, Muscarinic M3/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
5.
Cell Signal ; 21(4): 609-21, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167486

ABSTRACT

Signaling by the B cell antigen receptor (BCR) is essential for B lymphocyte homeostasis and immune function. In immature B cells, ligation of the BCR promotes growth arrest and apoptosis, and BCR-driven balancing between pro-apoptotic extracellular signal-regulated kinase 1 and 2 (ERK1/2) and anti-apoptotic phosphoinositide 3-kinase-dependent Akt seems to define the final cellular apoptotic response. Dysfunction of these late BCR signaling events can lead to the development of immunological diseases. Here we report on novel cyclic AMP-dependent mechanisms of BCR-induced growth arrest and apoptosis in the immature B lymphoma cell line WEHI-231. BCR signaling to ERK1/2 and Akt requires cyclic AMP-regulated Epac, the latter acting as a guanine nucleotide exchange factor for Rap1 and H-Ras independent of protein kinase A. Importantly, activation of endogenously expressed Epac by a specific cyclic AMP analog enhanced the induction of growth arrest (reduced DNA synthesis) and apoptosis (nuclear condensation, annexin V binding, caspase-3 cleavage and poly-ADP-ribose polymerase processing) by the BCR. Our data indicate that cyclic AMP-dependent Epac signals to ERK1/2 and Akt upon activation of Rap1 and H-Ras, and is involved in BCR-induced growth arrest and apoptosis in WEHI-231 cells.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Cyclic AMP/physiology , Guanine Nucleotide Exchange Factors/physiology , Lymphoma, B-Cell/pathology , Receptors, Antigen, B-Cell/physiology , Signal Transduction/physiology , Adenylyl Cyclase Inhibitors , Animals , Bacterial Toxins/pharmacology , Cell Division/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Enzyme Activation , Mice , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins p21(ras)/physiology , rap1 GTP-Binding Proteins/physiology
6.
J Invest Dermatol ; 128(6): 1487-98, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18172456

ABSTRACT

The lysophospholipids, sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA), stimulate chemotaxis and induce differentiation of human keratinocytes. As Ca(2+) plays an important role in keratinocyte differentiation, we studied Ca(2+) signaling by S1P and LPA in these cells, known to express mRNA transcripts of the S1P(1-5) and LPA(1-3) receptors, and the receptor subtypes involved in this process. S1P and LPA caused transient increases in intracellular free Ca(2+) concentration ([Ca(2+)](i)), with pEC(50) values of 8.5+/-0.11 and 7.5+/-0.23, respectively. The [Ca(2+)](i) increases are apparently mediated by stimulation of phospholipase C and involve Ca(2+) mobilization from thapsigargin-sensitive stores and subsequent Ca(2+) influx. The LPA-induced [Ca(2+)](i) increases were not inhibited by the LPA(1/3) receptor antagonist, dioctanoylglycerol pyrophosphate. The S1P-induced [Ca(2+)](i) increases were largely inhibited by the putative S1P(3) antagonist, BML-241, and the S1P(1/3) antagonist, VPC23019. The S1P(1)-specific agonist, SEW2871, did not increase [Ca(2+)](i) but stimulated chemotaxis of keratinocytes, which was fully blocked by S1P(1) antisense oligonucleotides. The data indicate that LPA and S1P potently increase [Ca(2+)](i) in human keratinocytes and that the effect of LPA is mediated by LPA(2), whereas that of S1P is mediated at least to a large part by S1P(3). The S1P(1) receptor, without stimulating [Ca(2+)](i) increases, mediates chemotaxis of keratinocytes.


Subject(s)
Calcium Signaling , Calcium/metabolism , Keratinocytes/cytology , Receptors, Lysophospholipid/metabolism , Cell Movement , Chemotaxis , Green Fluorescent Proteins/metabolism , Humans , Keratinocytes/metabolism , Lysophospholipids/metabolism , Models, Biological , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thapsigargin/metabolism , Thiazolidines/pharmacology , Type C Phospholipases/metabolism
7.
EMBO J ; 26(19): 4189-202, 2007 Oct 03.
Article in English | MEDLINE | ID: mdl-17853892

ABSTRACT

The activity state of cofilin, which controls actin dynamics, is driven by a phosphorylation-dephosphorylation cycle. Phosphorylation of cofilin by LIM-kinases results in its inactivation, a process supported by 14-3-3zeta and reversed by dephosphorylation by slingshot phosphatases. Here we report on a novel cellular function for the phosphorylation-dephosphorylation cycle of cofilin. We demonstrate that muscarinic receptor-mediated stimulation of phospholipase D1 (PLD1) is controlled by LIM-kinase, slingshot phosphatase as well as 14-3-3zeta, and requires phosphorylatable cofilin. Cofilin directly and specifically interacts with PLD1 and upon phosphorylation by LIM-kinase1, stimulates PLD1 activity, an effect mimicked by phosphorylation-mimic cofilin mutants. The interaction of cofilin with PLD1 is under receptor control and encompasses a PLD1-specific fragment (aa 585-712). Expression of this fragment suppresses receptor-induced cofilin-PLD1 interaction as well as PLD stimulation and actin stress fiber formation. These data indicate that till now designated inactive phospho-cofilin exhibits an active cellular function, and suggest that phospho-cofilin by its stimulatory effect on PLD1 may control a large variety of cellular functions.


Subject(s)
Nuclear Proteins/metabolism , Phospholipase D/metabolism , Protein Processing, Post-Translational/physiology , Receptors, Muscarinic/metabolism , 14-3-3 Proteins , Actins/genetics , Actins/metabolism , Cell Line , Gene Expression , Humans , Lim Kinases , Mutation , Nuclear Proteins/genetics , Phospholipase D/genetics , Phosphorylation , Protein Binding/physiology , Protein Kinases/genetics , Protein Kinases/metabolism , Receptors, Muscarinic/genetics
8.
Cell Signal ; 19(6): 1229-37, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17300916

ABSTRACT

The small GTPases, Rac1 and RhoA, are pivotal regulators of several essential, but distinct cellular processes. Numerous G-protein-coupled receptors signal to these GTPases, but with different specificities. Specifically, Gi-coupled receptors (GiPCRs) are generally believed to activate Rac1, but not RhoA, a process involving Gbetagamma-dimers and phosphatidylinositol 3-kinase (PI3K). Here we show that, depending on the expression level of the 519 amino acid isoform of regulator of G-protein signalling 3 (RGS3L), prototypical GiPCRs, like M2 muscarinic, A1 adenosine, and alpha2-adrenergic receptors, activate either Rac1 or RhoA in human embryonic kidney cells and neonatal rat cardiomyocyte-derived H10 cells. The switch from Rac1 to RhoA activation in H10 cells was controlled by fibroblast growth factor-2 (FGF-2), lowering the expression of RGS3L. Activation of both, Rac1 and RhoA, seen at low and high expression levels of RGS3L, respectively, was sensitive to pertussis toxin and the PI3K inhibitor LY294002 and mediated by Gbetagamma-dimers. We conclude that RGS3L functions as a molecular switch, redirecting GiPCRs via Gbetagamma-dimers and PI3K from Rac1 to RhoA activation. Considering the essential roles of Rac1 and RhoA in many signalling pathways, this additional function of RGS3L indicates a specific role of this protein in cellular signalling networks.


Subject(s)
GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Alanine/genetics , Animals , Asparagine/genetics , Carbachol/pharmacology , Cell Line , Dimerization , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/pharmacology , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Humans , Mutant Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RGS Proteins , RNA, Small Interfering/metabolism , Rats , Serum Response Factor/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism
9.
Curr Opin Pharmacol ; 7(2): 193-200, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17276729

ABSTRACT

Exchange protein activated by cyclic AMP (Epac) -- a cyclic AMP-activated guanine nucleotide exchange factor for Ras-like GTPases -- has emerged as a novel mediator of pivotal processes in the cardiovascular system, including cellular calcium handling, hypertrophy, integrin-mediated cell adhesion, establishment of cell polarity, cell migration and endothelial barrier functioning. Epac controls these various cellular responses apparently by signaling to several effector proteins. Spatiotemporal dynamics in the subcellular distribution of Epac-driven signaling networks probably determine the net outcome of cyclic AMP signaling in the cardiovascular system.


Subject(s)
Cardiovascular Physiological Phenomena , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Animals , Blood Cells/physiology , Blood Vessels/physiology , Cell Physiological Phenomena , Humans
10.
Biochim Biophys Acta ; 1768(4): 888-900, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17054901

ABSTRACT

G protein-coupled receptors (GPCRs) control a variety of fundamental cellular processes by regulating phospholipid signaling pathways. Essential for signaling by a large number of receptors is the hydrolysis of the membrane phosphoinositide PIP(2) by phospholipase C (PLC) into the second messengers IP(3) and DAG. Many receptors also stimulate phospholipase D (PLD), leading to the generation of the versatile lipid, phosphatidic acid. Particular PLC and PLD isoforms take differential positions in receptor signaling and are additionally regulated by small GTPases of the Ras, Rho and ARF families. It is now recognized that the PLC substrate, PIP(2), has signaling capacity by itself and can, by direct interaction, affect the activity and subcellular localization of PLD and several other proteins. As expected, the synthesis of PIP(2) by phosphoinositide 5-kinases is tightly regulated as well. In this review, we present an overview of how these signaling pathways are governed by GPCRs, explain the molecular basis for the spatially and temporally organized, highly dynamic quality of phospholipid signaling, and point to the functional connection of the pathways.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/biosynthesis , Phospholipase D/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Humans , Phospholipase D/genetics , Phospholipids/metabolism , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/genetics , Substrate Specificity
11.
Biochim Biophys Acta ; 1768(4): 923-40, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17078925

ABSTRACT

The lysophospholipids, sphingosine-1-phosphate (S1P), lysophosphatidic acid (LPA), sphingosylphosphorylcholine (SPC) and lysophosphatidylcholine (LPC), activate diverse groups of G-protein-coupled receptors that are widely expressed and regulate decisive cellular functions. Receptors of the endothelial differentiation gene family are activated by S1P (S1P(1-5)) or LPA (LPA(1-3)); two more distantly related receptors are activated by LPA (LPA(4/5)); the GPR(3/6/12) receptors have a high constitutive activity but are further activated by S1P and/or SPC; and receptors of the OGR1 cluster (OGR1, GPR4, G2A, TDAG8) appear to be activated by SPC, LPC, psychosine and/or protons. G-protein-coupled lysophospholipid receptors regulate cellular Ca(2+) homoeostasis and the cytoskeleton, proliferation and survival, migration and adhesion. They have been implicated in development, regulation of the cardiovascular, immune and nervous systems, inflammation, arteriosclerosis and cancer. The availability of S1P and LPA at their G-protein-coupled receptors is regulated by enzymes that generate or metabolize these lysophospholipids, and localization plays an important role in this process. Besides FTY720, which is phosphorylated by sphingosine kinase-2 and then acts on four of the five S1P receptors of the endothelial differentiation gene family, other compounds have been identified that interact with more ore less selectivity with lysophospholipid receptors.


Subject(s)
Lysophospholipids/metabolism , Receptors, Lysophospholipid/drug effects , Receptors, Lysophospholipid/metabolism , Signal Transduction , Animals , Disease/etiology , Humans , Receptors, Lysophospholipid/genetics
12.
J Biol Chem ; 281(31): 21837-21847, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16754664

ABSTRACT

The activation of the Ras-related GTPase R-Ras, which has been implicated in the regulation of various cellular functions, by G protein-coupled receptors (GPCRs) was studied in HEK-293 cells stably expressing the M3 muscarinic acetylcholine receptor (mAChR), which can couple to several types of heterotrimeric G proteins. Activation of the receptor induced a very rapid and transient activation of R-Ras. Studies with inhibitors and activators of various signaling pathways indicated that R-Ras activation by the M3 mAChR is dependent on cyclic AMP formation but is independent of protein kinase A. Similar to the rather promiscuous M3 mAChR, two typical G(s)-coupled receptors also induced R-Ras activation. The receptor actions were mimicked by an Epac-specific cyclic AMP analog and suppressed by depletion of endogenous Epac1 by small interfering RNAs, as well as expression of a cyclic AMP binding-deficient Epac1 mutant, but not by expression of dominant negative Rap GTPases. In vitro studies demonstrated that Epac1 directly interacts with R-Ras and catalyzes GDP/GTP exchange at this GTPase. Finally, it is shown that the cyclic AMP- and Epac-activated R-Ras plays a major role in the M3 mAChR-mediated stimulation of phospholipase D but not phospholipase C. Collectively, our data indicate that GPCRs rapidly activate R-Ras, that R-Ras activation by the GPCRs is apparently directly induced by cyclic AMP-regulated Epac proteins, and that activated R-Ras specifically controls GPCR-mediated phospholipase D stimulation.


Subject(s)
Cyclic AMP/metabolism , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phospholipase D/metabolism , Receptors, G-Protein-Coupled/metabolism , ras Proteins/metabolism , Catalysis , Cell Line , Guanosine Diphosphate/metabolism , Humans , Receptor, Muscarinic M3/metabolism , Signal Transduction
13.
J Lipid Res ; 47(8): 1762-70, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16687663

ABSTRACT

Olive oil consumption leads to high monounsaturated fatty acid intake, especially oleic acid, and has been associated with a reduced risk of hypertension. However, the molecular mechanisms and contribution of its different components to lower blood pressure (BP) require further evaluation. Here, we examined whether a synthetic, non-beta-oxidation-metabolizable derivative of oleic acid, 2-hydroxyoleic acid (2-OHOA), can normalize BP in adult spontaneously hypertensive rats (SHRs) and whether its antihypertensive action involves cAMP-dependent protein kinase A (PKA) and Rho kinase, two major regulators of vascular smooth muscle contraction. Oral administration of 2-OHOA to SHRs induced sustained systolic BP decreases in a time-dependent (1-7 days) and dose-dependent (100-900 mg/kg every 12 h) manner. After 7 days of treatment with 2-OHOA (600 mg/kg), the systolic BP of SHRs was similar to that of normotensive Wistar Kyoto rats, returning to its initial hypertensive level after withdrawal of 2-OHOA. This treatment strongly increased the protein expression of the catalytic and regulatory RIalpha and RIIalpha PKA subunits as well as PKA activity in aortas from SHRs. Consistently, administration of the PKA inhibitor 8-bromo adenosine-3',5'-cyclic monophosphorothioate, Rp isomer, to 2-OHOA-treated SHRs induced a pronounced reversal (up to 59%) of the antihypertensive effect of 2-OHOA. Additionally, 2-OHOA completely reversed the pathological overexpression of aortic Rho kinase found in SHRs, suppressing the vasoconstrictory Rho kinase pathway.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Oleic Acids/pharmacology , Protein Serine-Threonine Kinases/metabolism , Adenylyl Cyclases/metabolism , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Immunoblotting , Intracellular Signaling Peptides and Proteins , Male , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , rho-Associated Kinases
14.
Gastroenterology ; 130(3): 838-54, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16530523

ABSTRACT

BACKGROUND & AIMS: Portal hypertension is associated with arterial hypotension and vascular hypocontractility, which persists despite elevated plasma levels of vasoconstrictors. We investigated the role of the RhoA/Rho-kinase pathway in vascular smooth muscle hypocontractility of rats with secondary biliary cirrhosis. METHODS: Aortic expressions of RhoA and Rho-kinase were analyzed in sham-operated and BDL rats by reverse-transcription polymerase chain reaction (RT-PCR) and immunoblots. Activation of aortic RhoA was examined by pull down of guanosine triphosphate (GTP)-RhoA and membrane translocation of RhoA. Rho-kinase activity was assessed as phosphorylation of its substrate, moesin. Contractility of isolated aortic rings was determined myographically. The hemodynamic effect of the Rho-kinase inhibitor (R)-(+)-trans-N-(4-pyridyl)-4-(1-aminoethyl)-cyclohexanecarboxamide (Y-27632) was determined in vivo by measuring changes in mean arterial pressure and systemic vascular resistance (SVR) (microspheres). RESULTS: Contraction of aortic rings from BDL rats was impaired in response to the alpha(1)-adrenergic receptor agonist methoxamine but not to high molar KCl. Aortic expression of RhoA was unchanged in cirrhotic rats, whereas Rho-kinase was down-regulated posttranscriptionally. Methoxamine-induced activation of RhoA as well as basal and methoxamine-induced phosphorylation of moesin were strongly reduced in aortas from cirrhotic rats. Aortic rings from cirrhotic rats precontracted with methoxamine showed an increased sensitivity to relaxation with Y-27632. The drop in SVR induced by Y-27632 was larger in cirrhotic rats than in sham-operated rats. CONCLUSIONS: An impaired vascular activation of RhoA and a down-regulation of Rho-kinase might contribute to vasodilation and vascular hypocontractility in BDL-induced cirrhosis.


Subject(s)
Liver Cirrhosis, Experimental/physiopathology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Vasoconstriction , Vasodilation , rhoA GTP-Binding Protein/physiology , Amides/pharmacology , Animals , Aorta/physiology , Blood Pressure/drug effects , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Microfilament Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Pyridines/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , rho-Associated Kinases , rhoA GTP-Binding Protein/genetics
15.
J Biol Chem ; 280(12): 11134-9, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15632174

ABSTRACT

The monomeric GTPase RhoA, which is a key regulator of numerous cellular processes, is activated by a variety of G protein-coupled receptors, through either G12 or G(q) family proteins. Here we report that p63RhoGEF, a recently identified RhoA-specific guanine nucleotide exchange factor, enhances the Rho-dependent gene transcription induced by agonist-stimulated G(q/11)-coupled receptors (M3-cholinoceptor, histamine H1 receptor) or GTPase-deficient mutants of G alpha(q) and G alpha11. We further demonstrate that active G alpha(q) or G alpha11, but not G alpha12 or G alpha13, strongly enhances p63RhoGEF-induced RhoA activation by direct protein-protein interaction with p63RhoGEF at its C-terminal half. Moreover, the activation of p63RhoGEF by G alpha(q/11) occurs independently of and in competition to the activation of the canonical G alpha(q/11) effector phospholipase C beta. Therefore, our results elucidate a new signaling pathway by which G alpha(q/11)-coupled receptors specifically induce Rho signaling through a direct interaction of activated G alpha(q/11) subunits with p63RhoGEF.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology , Animals , Binding Sites , COS Cells , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Humans , Isoenzymes/metabolism , Phospholipase C beta , Rho Guanine Nucleotide Exchange Factors , Transcription, Genetic , Type C Phospholipases/metabolism
16.
Eur J Pharmacol ; 500(1-3): 87-99, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15464023

ABSTRACT

The membrane phospholipid, phosphatidylinositol 4,5-bisphosphate (PIP(2)), plays a critical role in various, apparently very different cellular processes. As precursor for second messengers generated by phospholipase C isoforms and class I phosphoinositide 3-kinases, PIP(2) is indispensable for cellular signaling by membrane receptors. In addition, PIP(2) directly affects the localization and activity of many cellular proteins via specific interaction with unique phosphoinositide-binding domains and thereby regulates actin cytoskeletal dynamics, vesicle trafficking, ion channel activity, gene expression and cell survival. The activity and subcellular localization of phosphatidylinositol 4-phosphate 5-kinase (PIP5K) isoforms, which catalyze the formation of PIP(2), are actively regulated by membrane receptors, by phosphorylation and by small GTPases of the Rho and ARF families. Spatially and temporally organized regulation of PIP(2) synthesis by PIP5K enables dynamic and versatile PIP(2) signaling and represents an important link in the execution of cellular tasks by Rho and ARF GTPases.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/physiology , Animals , GTP Phosphohydrolases/physiology , Humans , Isoenzymes/metabolism , Isoenzymes/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction
18.
J Biol Chem ; 279(45): 46497-508, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15319437

ABSTRACT

We have recently reported that two typical Gs-coupled receptors, the beta2-adrenergic receptor and the receptor for prostaglandin E1, stimulate phospholipase C-epsilon (PLC-epsilon) and increase intracellular Ca2+ concentration ([Ca2+]i) in HEK-293 cells and N1E-115 neuroblastoma cells, respectively, by a pathway involving Epac1, a cAMP-activated and Rap-specific guanine nucleotide exchange factor (GEF), and the GTPase Rap2B. Here we have demonstrated that these Gs-coupled receptors use this pathway to activate H-Ras and the extracellular signal-regulated kinases 1 and 2 (ERK1/2). Specifically, agonist activation of the receptors resulted in activation of H-Ras and ERK1/2. The latter action was suppressed by dominant negative H-Ras, but not Rap1A. The receptor actions were independent of protein kinase A but fully mimicked by an Epac-specific cAMP analog as well as by a constitutively active Rap2B mutant. On the other hand, a cAMP-binding-deficient Epac1 mutant, the Rap GTPase-activating proteinII, and a dominant negative Rap2B mutant suppressed receptor- and Epac-mediated activation of H-Ras and ERK1/2. Finally, we have demonstrated that activation of H-Ras and ERK1/2 requires the lipase activity of PLC-epsilon and the subsequent [Ca2+]i increase, suggesting that H-Ras activation is mediated by a Ca2+ -activated GEF. In line with this hypothesis, receptor-mediated activation of H-Ras and ERK1/2 was strongly enhanced by expression of RasGRP1, a Ca2+ -regulated Ras-GEF. Collectively, our data indicated that Gs-coupled receptors can activate H-Ras and subsequently the mitogen-activated protein kinases ERK1/2 by a Ca2+ -activated Ras-GEF, possibly RasGRP1, mediated by cAMP-activated Epac proteins, which then lead via Rap2B and PLC-epsilon stimulation to [Ca2+]i increase.


Subject(s)
Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanine Nucleotide Exchange Factors/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , ras Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunoblotting , MAP Kinase Signaling System , Phosphoinositide Phospholipase C , Plasmids/metabolism , Rats , Time Factors , Transfection , Type C Phospholipases/metabolism
19.
Mol Cell Biol ; 24(11): 4664-76, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15143162

ABSTRACT

Receptor tyrosine kinase regulation of phospholipase C-epsilon (PLC-epsilon), which is under the control of Ras-like and Rho GTPases, was studied with HEK-293 cells endogenously expressing PLC-coupled epidermal growth factor (EGF) receptors. PLC and Ca(2+) signaling by the EGF receptor, which activated both PLC-gamma1 and PLC-epsilon, was specifically suppressed by inactivation of Ras-related GTPases with clostridial toxins and expression of dominant-negative Rap2B. EGF induced rapid and sustained GTP loading of Rap2B, binding of Rap2B to PLC-epsilon, and Rap2B-dependent translocation of PLC-epsilon to the plasma membrane. GTP loading of Rap2B by EGF was inhibited by chelation of intracellular Ca(2+) and expression of lipase-inactive PLC-gamma1 but not of PLC-epsilon. Expression of RasGRP3, a Ca(2+)/diacylglycerol-regulated guanine nucleotide exchange factor for Ras-like GTPases, but not expression of various other exchange factors enhanced GTP loading of Rap2B and PLC/Ca(2+) signaling by the EGF receptor. EGF induced tyrosine phosphorylation of RasGRP3, but not RasGRP1, apparently caused by c-Src; inhibition of c-Src interfered with EGF-induced Rap2B activation and PLC stimulation. Collectively, these data suggest that the EGF receptor triggers activation of Rap2B via PLC-gamma1 activation and tyrosine phosphorylation of RasGRP3 by c-Src, finally resulting in stimulation of PLC-epsilon.


Subject(s)
ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Type C Phospholipases/metabolism , rap GTP-Binding Proteins/metabolism , CSK Tyrosine-Protein Kinase , Humans , Phosphoinositide Phospholipase C , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Time Factors , ras Guanine Nucleotide Exchange Factors , src-Family Kinases
20.
Cell Signal ; 16(8): 921-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15157671

ABSTRACT

We recently reported that several Gs-coupled receptors stimulate phospholipase C (PLC)-epsilon via increased formation of cyclic AMP and subsequent activation of the small GTPase Rap2B by the cyclic AMP-activated exchange factor Epac1. Here we show by studies in HEK-293 and N1E-115 neuroblastoma cells that this stimulation induced by Gs-coupled receptors or the direct adenylyl cyclase activator, forskolin, is potently inhibited by Gi-coupled receptors, known to inhibit cyclic AMP formation. PLC inhibition by the overexpressed M2 muscarinic receptor and the endogenously expressed sphingosine-1-phosphate and delta-opioid receptors was fully pertussis toxin-sensitive and accompanied by a reduction in Rap2B activation induced by Gs-coupled receptors. In contrast, Rap2B activation and PLC stimulation induced by membrane-permeable cyclic AMP analogues, including an Epac-specific activator, or PLC stimulation caused by constitutively active Rap2B were not affected by the Gi-coupled receptors. In summary, our data indicate that Gi-coupled receptors can inhibit PLC-epsilon, most likely by suppressing formation of cyclic AMP required for Epac-mediated Rap2B activation.


Subject(s)
Cyclic AMP/metabolism , Lysophospholipids/metabolism , Receptors, Opioid, delta/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Type C Phospholipases/antagonists & inhibitors , Cells, Cultured , Colforsin/pharmacology , Enzyme Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Pertussis Toxin/pharmacology , Phosphoinositide Phospholipase C , Receptors, Muscarinic/metabolism , Receptors, Opioid, delta/antagonists & inhibitors , Signal Transduction/drug effects , rap GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...