Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 283(1): 158-63, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11322783

ABSTRACT

Growth factor-induced activation of Akt (protein kinase B) is implicated in the proliferation of vascular smooth muscle cells (VSMC) in addition to antiapoptotic signaling. Although previous studies have documented increases in total Akt or Akt-1 activity in rodent VSMC, little is known about the regulation of Akt-2 or Akt-3 kinase activity in VSMC from any species. In the present study, reverse transcriptase-polymerase chain reaction revealed the expression of all three Akt isoforms in human aortic VSMC. In vitro kinase assays using immunoprecipitated Akt isoforms showed robust increases in Akt-3 activity after stimulation of human aortic VSMC with platelet-derived growth factor (PDGF), insulin, and insulin-like growth factor-1. In contrast, these growth factors produced modest and marginal increases in Akt-1 and Akt-2 kinase activity, respectively. Pretreatment of VSMC with a phosphoinositide-3kinase (PI-3K) inhibitor, LY294002, led to significant inhibition of growth factor(s)-induced increases in Akt-3 activity and DNA synthesis. The present findings provide the first direct evidence that the Akt-3 isoform is predominantly activated in human aortic VSMC. Moreover, these data suggest that PI-3K-dependent activation of Akt-3 may play a major role in VSMC proliferation.


Subject(s)
Aorta , Muscle, Smooth, Vascular/enzymology , Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Cells, Cultured , Coronary Vessels , DNA/biosynthesis , Enzyme Inhibitors/pharmacology , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oncogene Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Platelet-Derived Growth Factor/pharmacology , Precipitin Tests , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
Circ Res ; 87(4): 282-8, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10948061

ABSTRACT

Neointimal hyperplasia at the site of surgical intervention is a common and deleterious complication of surgery for cardiovascular diseases. We hypothesized that direct delivery of a cell-permeable growth-arresting lipid via the balloon tip of an embolectomy catheter would limit neointimal hyperplasia after stretch injury. We have previously demonstrated that sphingolipid-derived ceramide arrested the growth of smooth muscle cell pericytes in vitro. Here, we show that ceramide-coated balloon catheters significantly reduced neointimal hyperplasia induced by balloon angioplasty in rabbit carotid arteries in vivo. This ceramide treatment decreased the number of vascular smooth muscle cells entering the cell cycle without inducing apoptosis. In situ autoradiographic studies demonstrated that inflating the balloon catheter forced cell-permeable ceramide into the intimal and medial layers of the artery. Intercalation of ceramide into vascular smooth muscle cells correlated with rapid inhibition of trauma-associated phosphorylation of extracellular signal-regulated kinase and protein kinase B. These studies demonstrate the utility of cell-permeable ceramide as a novel therapy for reducing neointimal hyperplasia after balloon angioplasty.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/pathology , Ceramides/pharmacology , Angioplasty, Balloon, Coronary/methods , Animals , Apoptosis/physiology , Carotid Artery Injuries/metabolism , Carotid Stenosis/drug therapy , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Disease Models, Animal , Hyperplasia , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Postoperative Complications/drug therapy , Postoperative Complications/pathology , Postoperative Complications/prevention & control , Rabbits , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
3.
Am J Physiol Cell Physiol ; 279(1): C225-35, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898734

ABSTRACT

In addition to the well-documented role of nitric oxide (NO) as a vasodilator, NO has also been implicated in vascular smooth muscle cell (VSMC) growth arrest. Signaling mechanisms responsible for growth factor receptor-mediated VSMC proliferation include the extracellular signal-regulated kinase (ERK) and possibly the protein kinase B (PKB) cascade. Thus the present study was designed to test the hypothesis that, in A7r5 vascular smooth muscle-derived cells, platelet-derived growth factor (PDGF)-induced activation of either ERK or PKB is regulated by NO, which then modulates cellular proliferation and/or apoptosis. PKB-alpha was the predominant isoform of PKB expressed in A7r5 cells and was also expressed in rabbit carotid arteries and aortae. Phosphorylation of PKB-alpha and ERK induced by PDGF-BB was maximal within 5-15 min in A7r5 cells. Preincubation of A7r5 cells with the NO donor S-nitroso-N-acetylpenicillamine (SNAP) resulted in a biphasic regulation of PDGF-stimulated PKB-alpha phosphorylation and bioactivity. Acute exposure to SNAP significantly augmented PDGF-induced activation of PKB-alpha, whereas prolonged incubation led to a marked diminution in PDGF-induced activation of PKB-alpha. In contrast, SNAP did not affect PDGF-induced activation of ERK at any time point. The cGMP-independent effects of SNAP on PDGF-induced activation of PKB-alpha were established with the use of an inhibitor of soluble guanylyl cyclase, ODQ, as well as a cell-permeable analog of cGMP, 8-bromo-cGMP. Prolonged treatment of A7r5 cells with SNAP led to a significant decrease in DNA synthesis without an appreciable increase in apoptosis. These data suggest that, after prolonged exposure to SNAP, NO selectively attenuates PDGF-induced increase in PKB-alpha activation, which in turn may contribute to diminished VSMC proliferation by mechanisms involving growth arrest but not apoptosis.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/physiology , Platelet-Derived Growth Factor/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Aorta/metabolism , Apoptosis/drug effects , Becaplermin , Blood Vessels/growth & development , Carotid Arteries/metabolism , Cell Line , Endothelin-1/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/metabolism , Muscle, Smooth, Vascular/cytology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-sis , Rabbits , Rats , S-Nitroso-N-Acetylpenicillamine
4.
J Mol Cell Cardiol ; 31(4): 799-808, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10329207

ABSTRACT

The role of guanosine 3',5'-cyclic monophosphate (cGMP) in the regulation of cardiac contractility remains controversial. The present study has examined the effects of high concentrations of the nitric oxide (NO) donors, S-nitroso-N-acetylpenicillamine (SNAP) and 1,1-diethyl-2-hydroxy-2-nitroso-hydrazine (DEA/NO), on cGMP levels and isoproterenol-induced increases in contractility in rat cardiomyocytes before and after selective inhibition of soluble guanylyl cyclase with 1 H -[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ). In control myocytes, 100 microm SNAP or 100 microm DEA/NO increased cGMP levels by more than 15-fold at 2 and 6 min and produced marked attenuations of isoproterenol-mediated increases in maximal cell shortening over the same time period. The NO donors had no significant effect on basal cell shortening (in the absence of isoproterenol). Pretreatment of myocytes with 25 microm ODQ for 30 min resulted in a complete blockade of the SNAP- or DEA/NO-induced increases in cGMP with no reversal of negative inotropy. ODQ did not affect basal contractility, basal cGMP levels or isoproterenol-induced increases in cell shortening. Furthermore, myocytes exposed to the cGMP analog, 8-bromo-cGMP (100 microm), did not exhibit significant differences in basal contractility or isoproterenol-induced increases in cell shortening. These results suggest that attenuation of cardiac contractility by NO donors in rat cardiomyocytes occurs by a mechanism independent of increases in cGMP levels.


Subject(s)
Cyclic GMP/metabolism , Hydrazines/pharmacology , Myocardial Contraction/drug effects , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Animals , Cardiotonic Agents/metabolism , Cardiotonic Agents/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Isoproterenol/metabolism , Isoproterenol/pharmacology , Male , Myocardial Contraction/physiology , Myocardium/metabolism , Nitrogen Oxides , Oxadiazoles/pharmacology , Oxidation-Reduction , Penicillamine/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Wistar
5.
Mol Cell Biochem ; 163-164: 305-18, 1996.
Article in English | MEDLINE | ID: mdl-8974070

ABSTRACT

To evaluate the effects of the in vivo endotoxin treatment of the rat on (1) the contractile responses in the subsequently isolated papillary muscle to adrenergic and cholinergic agonists and (2) the biochemical parameters (cyclic GMP, nitric oxide synthesis, protein phosphorylation and ADP-ribosyslation) in the subsequently isolated cardiomyocytes. Following the in vivo endotoxin treatment (4 mg/kg i.p., 18 h), contractile responses to increasing amounts of isoprenaline or to increasing amounts of oxotremorine in the presence of a fixed amount of isoprenaline were determined in isolated papillary strips. Activities of nitric oxide synthase, guanylyl cyclase, as well as phosphorylation of phospholamban and troponin-inhibitory subunit, and pertussis toxin-catalyzed and endogenous ADP-ribosylations were determined in the intact cardiomyocytes and subcellular fractions. The increase in the force of contraction by isoprenaline was reduced, while its inhibition by oxotremorine was greater in the endotoxin-treated papillary strips. The activities of both nitric oxide synthase, primarily of the inducible form of the enzyme, and cytosolic guanylyl cyclase were higher while the phosphorylations of both phospholamban and troponin-inhibitory subunit were of lesser magnitude in the cardiomyocytes following the in vivo endotoxin treatment. Pertussis toxin-catalyzed ADP-ribosylation of the 41 kDa polypeptide, which is the alpha subunit of Gi, was also decreased. The results of the present study support the postulate that alterations in both the cyclic AMP and cyclic GMP signalling cascade contribute to the myocardial dysfunction caused by endotoxin and cytokines.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Cyclic GMP/biosynthesis , Heart/drug effects , Myocardium/metabolism , Nitric Oxide/biosynthesis , Pertussis Toxin , Virulence Factors, Bordetella/pharmacology , Animals , Calcium/metabolism , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Homeostasis , Male , Myocardial Contraction , Nitric Oxide Synthase/metabolism , Phosphorylation , Rats , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Receptors, Muscarinic/metabolism
6.
Brain Res ; 735(2): 311-3, 1996 Oct 07.
Article in English | MEDLINE | ID: mdl-8911670

ABSTRACT

Nitric oxide synthase (NOS) activity was examined in forebrain, cerebellum and optic lobes of adult domestic fowl, having a hereditary primary generalized convulsive disorder. NOS was approximately 2-fold higher in only the forebrain of adult epileptic fowl compared to non-epileptic (carrier) hatchmates. A significant increase in NOS was also evident in forebrains of 1-day-old epileptic chicks. Ca(2+)-dependency experiments confirmed that these increments were principally due to type I NOS (NOS-I). Induction of convulsions by intermittent photic stimulation did not affect pre-existing forebrain NOS-I activity. The present data suggest that an enhanced NO signaling may ensue in selected regions of the brain as an adaptive response to hereditary epileptogenesis.


Subject(s)
Epilepsy/enzymology , Nitric Oxide Synthase/metabolism , Poultry/metabolism , Prosencephalon/enzymology , Animals , Animals, Newborn/metabolism , Epilepsy/genetics , Male , Photic Stimulation , Reference Values
7.
Life Sci ; 56(7): PL169-74, 1995.
Article in English | MEDLINE | ID: mdl-7869826

ABSTRACT

Although several studies have shown that vanadate evokes vasoconstriction whether it elevates cytosolic free calcium, [Ca2+]i, in vascular smooth muscle (VSM) cells has not been investigated. The present study shows that acute additions of low concentrations of vanadate (10-200 microM) to cultured aortic smooth muscle cells (ASMC) produced a rapid and a concentration-dependent increase in [Ca2+]i with an EC50 (mean +/- SEM) value of 42 +/- 11 microM. Inclusion of vanadate (200 microM) led to a significant increase (p < 0.05) in the peak [Ca2+]i level to 190 +/- 23 nM from a basal level of 102 +/- 2 nM. At concentrations > 200 microM, vanadate caused quenching of fura-2 fluorescence. For example, addition of 1 mM vanadate led to an apparent decrease in fluorescence by about 50% (due to a quenching effect), followed by a transient rise. H2O2, which is used in the preparation of peroxide forms of vanadate, pervanadate (PV), also produced a rise in [Ca2+]i. These data suggest that vanadate promotes vascular tone by elevating [Ca2+]i in ASMC. However, [Ca2+]i measurements made with higher concentrations of vanadate and PV, using the fura-2 method, must be interpreted with caution.


Subject(s)
Calcium/metabolism , Muscle, Smooth, Vascular/drug effects , Vanadates/pharmacology , Animals , Aorta/metabolism , Cells, Cultured , Cytosol/metabolism , Dose-Response Relationship, Drug , Fluorescence , Hydrogen Peroxide/pharmacology , Male , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley
8.
Diabetes ; 43(8): 1033-40, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8039597

ABSTRACT

Incubation of cultured rat aortic smooth muscle cells (ASMCs) in a medium containing high glucose concentrations (25 mM) did not affect the basal cytosolic free calcium ([Ca2+]i) but led to significant reductions in peak [Ca2+]i response evoked by arginine vasopressin, angiotensin II, and endothelin-1 (ET-1). This was observed in both the presence and absence of extracellular Ca2+. Maintenance of rat ASMCs in a medium containing mannose (an osmotic control for high glucose) did not affect either the basal or peptide agonist-evoked increase in [Ca2+]i. However, pretreatment with either the nonselective protein kinase C (PKC) inhibitor staurosporine or the selective PKC inhibitor 2,6-diamino-N-([1-(1-oxotridecyl)-2 piperidinyl] methyl) hexanamide reversed the attenuating effect of high glucose on peak [Ca2+]i response evoked by ET-1. Also, short-term incubation of ASMCs with the active phorbol ester, phorbol 12-myristate 13-acetate, led to a reduction in peak [Ca2+]i response to all three agonists, whereas the inactive phorbol ester, 4 alpha-phorbol 12,13-didecanoate, which does not activate PKC, had no such effect. Although high-glucose treatment of rat ASMCs led to significant reductions in the maximal number of binding sites to the extent of 39% of [125I]ET-1 specific binding, no significant differences in the affinity (Kd approximately 110 pM) characteristics were evident between control and high-glucose treatment groups. It is proposed that incubation of rat ASMCs with high glucose enhances the de novo synthesis of diacylglycerol and activates membrane-bound PKC and that this, in turn, impairs agonist-mediated intracellular Ca2+ mobilization.


Subject(s)
Angiotensin II/pharmacology , Arginine Vasopressin/pharmacology , Cytosol/metabolism , Endothelins/pharmacology , Glucose/pharmacology , Muscle, Smooth, Vascular/metabolism , Alkaloids/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Calcium/pharmacology , Cells, Cultured , Culture Media , Endothelins/metabolism , Glucose/administration & dosage , Male , Mannose/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Piperidines/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Staurosporine , Tetradecanoylphorbol Acetate/pharmacology
10.
Br J Pharmacol ; 108(1): 6-8, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8428214

ABSTRACT

Primary cultures of neonatal rat cardiomyocytes were pretreated for 16 h with either nonselective (staurosporine, 100 nM) or selective (NPC15437, 20 microM) protein kinase C (PKC) inhibitors. These inhibitors did not affect the basal cytosolic free calcium, [Ca2+]i, level (106 +/- 12 nM) as determined by fura-2 fluorescence methodology. Both agents significantly enhanced the maximal [Ca2+]i responses to endothelin-1 (ET-1) and attenuated the peak [Ca2+]i responses to arginine vasopressin and angiotensin II. They did not alter the EC50 values of any of these agonists. Since depletion of [Ca2+]o led to only partial attenuation of the enhanced response to ET-1 in the treatment groups, it is likely that PKC inhibition results in an exaggerated intracellular mobilization of Ca2+ to ET-1. It is concluded that PKC modulates agonist(s)-evoked intracellular Ca2+ mobilization and that the nature of regulation is governed by the agonist.


Subject(s)
Angiotensin II/pharmacology , Arginine Vasopressin/pharmacology , Calcium/metabolism , Endothelins/pharmacology , Myocardium/metabolism , Protein Kinase C/antagonists & inhibitors , Alkaloids/pharmacology , Animals , Cells, Cultured , Cytosol/metabolism , Fura-2 , Myocardium/cytology , Piperidines/pharmacology , Rats , Staurosporine
SELECTION OF CITATIONS
SEARCH DETAIL
...