Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 83(1): 123-30, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19377066

ABSTRACT

AIMS: Acute administration of aspirin increases nitric oxide (NO) synthesis by platelets, an effect not shared by other non-steroidal anti-inflammatory drugs. The aim of the present study was to determine the mechanism by which aspirin acutely increases the activity of NO synthase type 3 (NOS-3), the predominant NOS isoform expressed by platelets, and specifically whether this occurs through an increase in its acetylation. METHODS AND RESULTS: Platelets isolated from the blood of healthy human subjects were exposed in vitro to vehicle or aspirin at different concentrations (5 micromol/L-4 mmol/L). Changes in intraplatelet Ca(2+) concentration were determined from fura-2 fluorescence. Following immunoprecipitation of NOS-3 from platelet lysates, its activity was determined from l-[(3)H]arginine to l-[(3)H]citrulline conversion, and its serine phosphorylation quantified by western blotting. Acetylation of NOS-3 in platelets was assessed by the incorporation of radioactivity into the immunoprecipitated enzyme from [acetyl-(14)C]aspirin. Following transfection of HeLa cells with NOS-3, NO biosynthesis in response to aspirin was determined from cyclic GMP measurement, and sites of NOS-3 acetylation were ascertained by liquid chromatography-tandem mass spectrometry. At all concentrations tested, aspirin increased the activity of NOS-3 from platelets. This was not associated with any measurable change in intraplatelet Ca(2+) concentration. Serine phosphorylation of NOS-3 in platelets was decreased, and this was especially marked for serine-1177 phosphorylation, whereas acetylation of NOS-3 was increased, by aspirin incubation. HeLa cells transfected with NOS-3 exhibited an increase in NO biosynthesis following aspirin exposure, and this was associated with acetylation of the enzyme on both serine-765 and serine-771. CONCLUSION: Aspirin acetylates NOS-3 acutely in platelets, and this causes an increase in its activity as well as a decrease in its phosphorylation. It is also possible that aspirin indirectly affects NOS-3 activity by acetylating other substrates within the platelet, but this remains to be determined.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Blood Platelets/enzymology , Nitric Oxide Synthase Type III/metabolism , Acetylation/drug effects , Adolescent , Adult , Blood Platelets/cytology , Blood Platelets/drug effects , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , HeLa Cells , Humans , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Serine/metabolism , Transfection , Young Adult
2.
Cardiovasc Res ; 75(4): 793-802, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17572401

ABSTRACT

OBJECTIVE: Ageing is associated with an increase in atherothrombotic disease. Platelet-derived nitric oxide (NO) inhibits platelet activation, but the effect of age on platelet NO signaling is unknown. We investigated platelet NO biosynthesis and responsiveness in older (> 45 years old) as compared with younger (< 30 years old) healthy human subjects. METHODS: Platelet NO synthase (NOS) activity was evaluated by l-[3H]-arginine to l-[3H]-citrulline conversion, and cGMP was determined by radioimmunoassay. Platelet expression of NOS3, phosphoserine-1177-NOS3 and soluble guanylyl cyclase (sGC) were quantified by Western blotting. Circulating monocyte-platelet aggregates (MPA) were measured by flow cytometry. RESULTS: Basal NOS activity was similar in both groups. By contrast, whereas both albuterol and collagen stimulated platelet NOS in younger subjects, stimulation was absent in older subjects. Platelet NOS3 expression was similar in both age groups, but NOS3 serine-1177 phosphorylation was greater in younger subjects. Basal, albuterol- and collagen-stimulated cGMP, as well as sGC expression, were all greater in younger than older subjects, and within the younger group both cGMP (basal and stimulated) and sGC expression were greater in women than in men. Circulating MPA were greater in older subjects and, whilst NOS inhibition increased MPA further in both groups, it did so to a lesser extent in the older age bracket. CONCLUSIONS: These data suggest that platelet NO production and responsiveness decrease with age, and this is reflected in increased circulating MPA.


Subject(s)
Aging/physiology , Blood Platelets/metabolism , Monocytes/pathology , Nitric Oxide/metabolism , Signal Transduction/physiology , Adult , Age Factors , Albuterol/pharmacology , Atherosclerosis/blood , Blood Platelets/drug effects , Blood Platelets/pathology , Blotting, Western , Calcium/analysis , Cell Adhesion , Collagen/pharmacology , Cyclic GMP/analysis , Cyclic GMP/metabolism , Female , Guanylate Cyclase/metabolism , Humans , Male , Middle Aged , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Platelet Adhesiveness , Risk Factors , Sex Factors , Stimulation, Chemical , Superoxides/analysis , Superoxides/metabolism , Thrombosis/blood
3.
Proc Natl Acad Sci U S A ; 104(21): 8839-44, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17502619

ABSTRACT

Cytoskeletal proteins are crucial in maintaining cellular structure and, in certain cell types, also play an essential role in motility and shape change. Nitric oxide (NO) is an important paracrine mediator of vascular and platelet function and is produced in the vasculature by the enzyme NO synthase type 3 (NOS-3). Here, we demonstrate in human platelets that the polymerization state of beta-actin crucially regulates the activation state of NOS-3, and hence NO formation, through altering its binding of heat shock protein 90 (Hsp90). We found that NOS-3 binds to the globular, but not the filamentous, form of beta-actin, and the affinity of NOS-3 for globular beta-actin is, in turn, increased by Hsp90. Formation of this ternary complex among NOS-3, globular beta-actin, and Hsp90, in turn, results in an increase in both NOS activity and cyclic guanosine-3',5'-monophosphate, an index of bioactive NO, as well as an increased rate of Hsp90 degradation, thus limiting the duration for which NOS-3 remains activated. These observations suggest that beta-actin plays a critical role in regulating NO formation and signaling in platelets.


Subject(s)
Actins/metabolism , Blood Platelets/metabolism , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Actins/chemistry , Amino Acid Sequence , Humans , Molecular Sequence Data , Nitric Oxide/metabolism , Protein Binding
4.
J Physiol ; 576(Pt 2): 585-94, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16873402

ABSTRACT

Endothelial beta(2)-adrenoceptor (beta(2)AR) stimulation increases nitric oxide (NO) generation, but the underlying cellular mechanisms are unclear. We examined the role of l-arginine transport and of phosphorylation of NO synthase 3 (NOS-3) in beta(2)AR-mediated NO biosynthesis by human umbilical vein endothelial cells (HUVEC). To this end, we assessed l-arginine uptake, NOS activity (from l-arginine to l-citrulline conversion), membrane potential (using [(3)H]tetraphenylphosphonium), as well as serine phosphorylation of NOS-3 (by Western blotting and mass spectrometry), in HUVEC treated with betaAR agonists or cyclic AMP-elevating agents. beta(2)AR stimulation increased l-arginine transport, as did cyclic AMP elevation with either forskolin or dibutyryl cyclic AMP, and this increase was inhibitable by N-ethylmaleimide. Blockade of l-arginine uptake by l-lysine inhibited NOS activity and, conversely, blockade of NOS using N(omega)-nitro-l-arginine methyl ester (l-NAME) inhibited l-arginine transport. beta(2)AR stimulation also caused a membrane hyperpolarization inhibitable by l-NAME, suggesting that the increase in l-arginine uptake occurred in response to NO-mediated hyperpolarization. beta(2)AR activation also increased NOS activity and phosphorylation of NOS-3 on serine-1177, and these increases were attenuated by inhibition of protein kinase A (PKA), phosphatidylinositol 3-kinase (PI3K) or Akt, and abolished by coinhibition of PKA and Akt. These findings suggest that beta(2)AR-mediated NOS-3 activation in HUVEC is mediated through phosphorylation of NOS-3 on serine-1177 through both the PKA and the PI3K/Akt systems, and is sustained by an increase in l-arginine uptake resulting from NO-mediated membrane hyperpolarization.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Receptors, Adrenergic, beta-2/metabolism , Umbilical Veins/cytology , Amino Acid Sequence , Arginine/metabolism , Biological Transport/physiology , Cells, Cultured , Cyclic AMP/metabolism , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Membrane Potentials , Molecular Sequence Data , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Receptors, Adrenergic, beta-2/genetics , Serine/metabolism
5.
Br J Pharmacol ; 143(3): 397-403, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15351777

ABSTRACT

Vasorelaxation to beta(2)-adrenoceptor stimulation occurs through both endothelium-dependent and endothelium-independent mechanisms, and the former is mediated through Ca(2+)-independent activation of endothelial-type nitric oxide synthase (NOS-3). Since Ca(2+)-independent NOS-3 activation may occur through its serine phosphorylation via protein kinase A (PKA) or Akt, we determined the PKA and Akt dependency of beta(2)-adrenergic relaxation of rat aorta. Rat aortic rings were pre-incubated with the PKA inhibitor H-89 (10(-7) m), the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin (5 x 10(-7) m), Akt inhibitor (10(-5) m), or vehicle, in the absence or presence of the NOS inhibitor N(G)-nitro-l-arginine methyl ester (l-NAME, 10(-4) m). Rings were then contracted with phenylephrine (10(-7) m), and concentration-relaxation responses determined to the beta(2)-adrenoceptor agonist albuterol. Rings exhibited a concentration-dependent relaxation to albuterol: pEC(50) 6.9+/-0.2, E(max) 88.2+/-4.0%. l-NAME attenuated E(max) to 60.2+/-3.5% (P<0.001). In the presence of l-NAME, wortmannin or Akt inhibitor did not influence albuterol responses, whereas H-89 reduced E(max) further, to 27.5+/-2.2% (P<0.001). In the absence of l-NAME, E(max) to albuterol was reduced by H-89, wortmannin or Akt inhibitor, to 56.2+/-2.2, 56.0+/-1.6 and 55.4+/-1.8%, respectively (P<0.001 for each); the combinations H-89 plus wortmannin or H-89 plus Akt inhibitor reduced E(max) further still. Western blotting of NOS-3 immunoprecipitates from rat aortas confirmed that albuterol increased serine phosphorylation of NOS-3, and this increase was attenuated by H-89 or Akt inhibitor. Our results indicate that beta(2)-adrenoceptor stimulation relaxes rat aorta through both NO-dependent and independent mechanisms. The latter is predominantly PKA-mediated, whereas the former occurs through both PKA and PI3K/Akt activation.


Subject(s)
Aorta, Thoracic/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Adrenergic, beta-2/physiology , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Albuterol/pharmacology , Androstadienes/pharmacology , Animals , Aorta, Thoracic/drug effects , Blotting, Western , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Propanolamines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Sulfonamides/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , Wortmannin
6.
Cardiovasc Res ; 59(1): 152-9, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12829186

ABSTRACT

OBJECTIVE: We examined the effects of aspirin on basal and beta-adrenoceptor (beta-AR)-mediated nitric oxide synthase (NOS) activity in normal platelets. METHODS: NOS activity was determined from the conversion of L-[3H]arginine to L-[3H]citrulline, both basally and following beta-AR stimulation, in platelets from healthy human subjects following both short- and long-term aspirin administration. RESULTS: Basal L-[3H]citrulline increased following aspirin 800 mg administered intravenously in vivo, from 0.31+/-0.12 to 0.76+/-0.14 pmol/10(8) platelets (P<0.01). Isoproterenol at 1 micromol/l increased platelet NOS activity before but not following intravenous aspirin. After short-term in vitro treatment with aspirin 10 micromol/l, 400 micromol/l or 4 mmol/l, basal platelet L-[3H]citrulline increased similarly, an effect not seen with indomethacin 100 micromol/l or ibuprofen 10 micromol/l. Platelet NOS activity was not increased by albuterol 1 micromol/l, in the presence of indomethacin, ibuprofen or aspirin in vitro. By contrast, oral aspirin 75 mg daily for 14 days did not affect basal platelet NOS activity, but abolished beta-adrenergic NOS activation. CONCLUSIONS: Aspirin activates basal platelet NOS acutely, but not chronically, through a mechanism independent of cyclooxygenase (COX) inhibition. By contrast, both short- and long-term aspirin treatment inhibit platelet beta-adrenergic NOS activation by a COX-dependent mechanism. This indicates that aspirin exerts divergent effects on basal and beta-AR-stimulated platelet NOS activity, which are likely to be of clinical relevance.


Subject(s)
Aspirin/administration & dosage , Blood Platelets/enzymology , Nitric Oxide Synthase/metabolism , Platelet Aggregation Inhibitors/administration & dosage , Adrenergic beta-Agonists/pharmacology , Adult , Albuterol/pharmacology , Analysis of Variance , Aspirin/therapeutic use , Blood Platelets/drug effects , Citrulline/metabolism , Cyclooxygenase Inhibitors/pharmacology , Drug Administration Schedule , Female , Humans , Indomethacin/pharmacology , Isoproterenol/pharmacology , Male , Middle Aged , Nitric Oxide Synthase/antagonists & inhibitors , Platelet Aggregation Inhibitors/therapeutic use , omega-N-Methylarginine/pharmacology
7.
Br J Pharmacol ; 136(3): 375-82, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12023940

ABSTRACT

1. We investigated the nitric oxide (NO) dependence of vasorelaxation in response to different calcium channel blockers (CCB), in rabbit femoral artery in vivo. 2. Anaesthetized rabbits underwent femoral artery ligation, and blood from the proximal artery was returned distal to the ligature through a constant infusion pump. The effects of local injection of CCB on perfusion pressure and plasma nitrite+nitrate (NO(x), which reflects local NO biosynthesis) concentration in this system were determined. 3. Intra-arterial verapamil, nifedipine or amlodipine 10 micromol x kg(-1) each reduced perfusion pressure. Pre-treatment with intra-arterial N(G)-nitro-L-arginine methyl ester (L-NAME, a NO synthase inhibitor) 1 micromol x kg(-1) did not affect responses to verapamil or nifedipine, but attenuated the reduction in perfusion pressure to amlodipine, from 33.2+/-2.1% to 22.5+/-1.6% (P=0.002). 4. Intra-arterial amlodipine--unlike verapamil or nifedipine--increased femoral venous NO(x), from 9.1+/-0.4 microM to 14.1+/-0.5 microM (P=0.005). 5. The bradykinin B2 receptor antagonist HOE 140, 30 mg x kg(-1), attenuated the reduction in perfusion pressure and abolished the rise in venous NO(x) concentration, following intra-arterial amlodipine. 6. Amlodipine potently inhibited serum angiotensin converting-enzyme (ACE) activity in vitro, as effectively as enalapril at similar concentrations. 7. These results suggest that the vasorelaxant effects of nifedipine and verapamil are NO-independent, whereas those of amlodipine are partly NO-dependent, in rabbit femoral artery in vivo. This effect of amlodipine occurs through B2 receptor activation, and may be related to an increase in local bradykinin through inhibition of ACE.


Subject(s)
Amlodipine/pharmacology , Calcium Channel Blockers/pharmacology , Femoral Artery/drug effects , Nifedipine/pharmacology , Nitric Oxide/metabolism , Receptors, Bradykinin/drug effects , Vasodilation/drug effects , Verapamil/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular/enzymology , Femoral Artery/physiology , Humans , Nitric Oxide Synthase/metabolism , Peptidyl-Dipeptidase A/blood , Rabbits , Receptor, Bradykinin B2
SELECTION OF CITATIONS
SEARCH DETAIL
...