Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 41(2): 683-697, 2021 02.
Article in English | MEDLINE | ID: mdl-33267663

ABSTRACT

OBJECTIVE: Using 3KO (triple NOX [NADPH oxidase] knockout) mice (ie, NOX1-/-/NOX2-/-/NOX4-/-), we aimed to clarify the role of this family of enzymes in the regulation of platelets in vitro and hemostasis in vivo. Approach and Results: 3KO mice displayed significantly reduced platelet superoxide radical generation, which was associated with impaired platelet aggregation, adhesion, and thrombus formation in response to the key agonists collagen and thrombin. A comparison with single-gene knockouts suggested that the phenotype of 3KO platelets is the combination of the effects of the genetic deletion of NOX1 and NOX2, while NOX4 does not show any significant function in platelet regulation. 3KO platelets displayed significantly higher levels of cGMP-a negative platelet regulator that activates PKG (protein kinase G). The inhibition of PKG substantially but only partially rescued the defective phenotype of 3KO platelets, which are responsive to both collagen and thrombin in the presence of the PKG inhibitors KT5823 or Rp-8-pCPT-cGMPs, but not in the presence of the NOS (NO synthase) inhibitor L-NG-monomethyl arginine. In vivo, triple NOX deficiency protected against ferric chloride-driven carotid artery thrombosis and experimental pulmonary embolism, while hemostasis tested in a tail-tip transection assay was not affected. Procoagulatory activity of platelets (ie, phosphatidylserine surface exposure) and the coagulation cascade in platelet-free plasma were normal. CONCLUSIONS: This study indicates that inhibiting NOXs has strong antithrombotic effects partially caused by increased intracellular cGMP but spares hemostasis. NOXs are, therefore, pharmacotherapeutic targets to develop new antithrombotic drugs without bleeding side effects.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Carotid Artery Thrombosis/enzymology , NADPH Oxidases/blood , Platelet Activation , Pulmonary Embolism/enzymology , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/genetics , Carotid Artery Thrombosis/prevention & control , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fibrinolytic Agents/pharmacology , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Platelet Activation/drug effects , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Signal Transduction , Superoxides/blood
2.
High Alt Med Biol ; 18(1): 27-36, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27740865

ABSTRACT

Padhy, Gayatri, Anamika Gangwar, Manish Sharma, Kalpana Bhargava, and Niroj Kumar Sethy. Plasma proteomics of Ladakhi natives reveal functional regulation between renin-angiotensin system and eNOS-cGMP pathway. High Alt Med Biol. 18:27-36, 2017.-Humans have been living in high altitudes for more than 25,000 years but the molecular pathways promoting survival and performance in these extreme environments are not well elucidated. In an attempt to understand human adaptation to high altitudes, we used two-dimensional gel electrophoresis combined with MALDI-TOF/TOF to identify plasma proteins and associated pathways of ethnic Ladakhi natives residing at 3520 m. This resulted in the identification of 36 differential proteins compared with sea-level individuals. Proteins belonging to coagulation cascade and complement activation were found to be less abundant in Ladakhi natives. Interestingly, we observed lower abundance of angiotensinogen (ANGT) and subsequent analysis also revealed lower levels of both ANGT and angiotensin II (Ang II) in Ladakhi natives. Concomitantly, we observed elevated levels of eNOS, phosphorylated eNOS (Ser1177), and plasma biomarkers for nitric oxide (NO) production (nitrate and nitrite) and availability (cGMP). These results suggest that functional interplay between renin-angiotensin system and NO-cGMP pathway contributes to the hypoxia adaptation in Ladakhi natives. These findings will augment the present understanding of higher NO and NO-derived metabolite availability during human adaptation to high altitude.


Subject(s)
Altitude , Blood Proteins/analysis , Cyclic GMP-Dependent Protein Kinases/blood , Nitric Oxide Synthase Type III/blood , Proteomics/methods , Renin-Angiotensin System/genetics , Acclimatization/genetics , Adult , Altitude Sickness/blood , Altitude Sickness/ethnology , Altitude Sickness/genetics , Blood Coagulation Factors/analysis , Complement Activation , Healthy Volunteers , Humans , India/ethnology , Male , Population Groups/ethnology , Population Groups/genetics , Signal Transduction/genetics , Young Adult
3.
Platelets ; 25(1): 1-7, 2014.
Article in English | MEDLINE | ID: mdl-23469931

ABSTRACT

Cyclic guanosine-3',5'-monophoshate (cGMP) is the common second messenger for the cardiovascular effects of nitric oxide (NO) and natriuretic peptides (NP; e.g. atrial NP [ANP]), which activate soluble and particulate guanylyl cyclases, respectively. The role of NO in regulating cGMP and platelet function is well documented, whereas there is little evidence supporting a role for NPs in regulating platelet reactivity. By studying platelet aggregation and secretion in response to a PAR-1 peptide, collagen and ADP, and phosphorylation of the cGMP-dependent protein kinase (PKG) substrate vasodilator-stimulated phosphoprotein (VASP) at serine 239, we evaluated the effects of NPs in the absence or presence of the non-selective cGMP and cAMP phosphodiesterase (PDE) inhibitor, 3-isobutyl-1-methylxanthine (IBMX). Our results show that NPs, possibly through the clearance receptor (natriuretic peptide receptor-C) expressed on platelet membranes, increase VASP phosphorylation but only following PDE inhibition, indicating a small, localised cGMP synthesis. As platelet aggregation and secretion measured under the same conditions were not affected, we conclude that the magnitude of PKG activation achieved by NPs in platelets per se is not sufficient to exert functional inhibition of platelet involvement in haemostasis.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Adhesion Molecules/blood , Microfilament Proteins/blood , Natriuretic Peptides/pharmacology , Phosphoproteins/blood , 1-Methyl-3-isobutylxanthine/pharmacology , Blood Platelets/enzymology , Cyclic GMP/biosynthesis , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Humans , Natriuretic Peptides/blood , Peptide Fragments/blood , Peptide Fragments/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Platelet Aggregation/drug effects
4.
Biochem Pharmacol ; 84(6): 793-803, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22750553

ABSTRACT

Activation of peroxisome proliferator-activated receptor (PPAR) isoforms (α, ß/δ, and γ) is known to inhibit platelet aggregation. In the present study, we examined whether PPARs-mediated pathways contribute to the antiplatelet activity of magnolol, a compound purified from Magnolia officinalis. Magnolol (20-60 µM) dose-dependently enhanced the activity and intracellular level of PPAR-ß/γ in platelets. In the presence of selective PPAR-ß antagonist (GSK0660) or PPAR-γ antagonist (GW9662), the inhibition of magnolol on collagen-induced platelet aggregation and intracellular Ca(2+) mobilization was significantly reversed. Moreover, magnolol-mediated up-regulation of NO/cyclic GMP/PKG pathway and Akt phosphorylation leading to increase of eNOS activity were markedly abolished by blocking PPAR-ß/γ activity. Additionally, magnolol significantly inhibited collagen-induced PKCα activation through a PPAR-ß/γ and PKCα interaction manner. The arachidonic acid (AA) or collagen-induced thromboxane B(2) formation and elevation of COX-1 activity caused by AA were also markedly attenuated by magnolol. However, these above effects of magnolol on platelet responses were strongly reduced by simultaneous addition of GSK0660 or GW9662, suggesting that PPAR-ß/γ-mediated processes may account for magnolol-regulated antiplatelet mechanisms. Similarly, administration of PPAR-ß/γ antagonists remarkably abolished the actions of magnolol in preventing platelet plug formation and prolonging bleeding time in mice. Taken together, we demonstrate for the first time that the antiplatelet and anti-thrombotic activities of magnolol are modulated by up-regulation of PPAR-ß/γ-dependent pathways.


Subject(s)
Biphenyl Compounds/pharmacology , Lignans/pharmacology , PPAR gamma/agonists , PPAR-beta/agonists , Platelet Aggregation Inhibitors/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Cyclic GMP/biosynthesis , Cyclic GMP-Dependent Protein Kinases/blood , Cyclooxygenase 1/blood , Fibrinolytic Agents/pharmacology , Guanylate Cyclase/blood , Mice , Mice, Inbred ICR , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/blood , PPAR gamma/antagonists & inhibitors , PPAR gamma/blood , PPAR-beta/antagonists & inhibitors , PPAR-beta/blood , Phosphorylation , Platelet Aggregation/drug effects , Protein Kinase C-alpha/blood , Proto-Oncogene Proteins c-akt/blood , Rabbits , Receptors, Cytoplasmic and Nuclear/blood , Signal Transduction , Soluble Guanylyl Cyclase , Thromboxane B2/blood , Up-Regulation
5.
J Thromb Haemost ; 10(2): 167-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22136590

ABSTRACT

Endothelial prostacyclin and nitric oxide potently inhibit platelet functions. Prostacyclin and nitric oxide actions are mediated by platelet adenylyl and guanylyl cyclases, which synthesize cyclic AMP (cAMP) and cyclic GMP (cGMP), respectively. Cyclic nucleotides stimulate cAMP-dependent protein kinase (protein kinase A [PKA]I and PKAII) and cGMP-dependent protein kinase (protein kinase G [PKG]I) to phosphorylate a broad panel of substrate proteins. Substrate phosphorylation results in the inactivation of small G-proteins of the Ras and Rho families, inhibition of the release of Ca(2+) from intracellular stores, and modulation of actin cytoskeleton dynamics. Thus, PKA/PKG substrates translate prostacyclin and nitric oxide signals into a block of platelet adhesion, granule release, and aggregation. cAMP and cGMP are degraded by phosphodiesterases, which might restrict signaling to specific subcellular compartments. An emerging principle of cyclic nucleotide signaling in platelets is the high degree of interconnection between activating and cAMP/cGMP-dependent inhibitory signaling pathways at all levels, including cAMP/cGMP synthesis and breakdown, and PKA/PKG-mediated substrate phosphorylation. Furthermore, defects in cAMP/cGMP pathways might contribute to platelet hyperreactivity in cardiovascular disease. This article focuses on recent insights into the regulation of the cAMP/cGMP signaling network and on new targets of PKA and PKG in platelets.


Subject(s)
Blood Platelets/metabolism , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic AMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Cyclic GMP/blood , Signal Transduction , Animals , Blood Platelets/drug effects , Humans , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/drug effects
6.
Cardiovasc Res ; 92(2): 328-37, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21859817

ABSTRACT

AIMS: We previously reported that C-reactive protein bioactivity on thrombogenesis was based on loss of its pentameric symmetry, resulting in formation of monomeric C-reactive protein. Our purpose was to provide mechanistic information on the direct effects of C-reactive protein isoforms on platelet activation and provide a C-reactive protein dissociation mechanism in circulating blood. METHODS AND RESULTS: C-reactive protein-induced platelet activation was evaluated by flow cytometry. Platelet aggregation, clot properties, and coagulation were also measured. Washed platelets were incubated with C-reactive protein isoforms and vasodilator-stimulated phosphoprotein (VASP) phosphorylation was analysed by western blot and immunofluorescence. C-reactive protein dissociation under flow was evaluated by confocal microscopy on the surface of adhered platelets after perfusing human blood containing pentameric C-reactive protein at different shear rates. Dissociated monomeric C-reactive protein thrombogenicity was measured in flow experiments. Platelet aggregation and flow cytometry analysis revealed that monomeric C-reactive protein significantly induced platelet aggregation, surface P-selectin and CD63 exposure, and glycoprotein IIb-IIIa activation, whereas pentameric C-reactive protein was unable to produce any effect. p38 mitogen-activated protein kinase (MAPK) and Jun N-terminal kinase (JNK) inhibitors, as well as CD36 blocking antibody partially inhibited monomeric C-reactive protein-induced platelet activation and aggregation. Additionally, monomeric C-reactive protein significantly induced VASP dephosphorylation at serine 239. We found that pentameric C-reactive protein dissociated into monomeric C-reactive protein on the surface of activated adhered platelets under flow conditions and that this generated monomeric C-reactive protein promoted further platelet recruitment. CONCLUSIONS: These data indicate that whereas serum pentameric C-reactive protein may not affect platelet activation, monomeric C-reactive protein, which dissociates from pentameric C-reactive protein on the surface of activated platelets, could contribute to atherothrombotic complications by promoting thrombosis.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , C-Reactive Protein/metabolism , Platelet Adhesiveness , Platelet Aggregation , Thrombosis/blood , Blotting, Western , C-Reactive Protein/chemistry , Cell Adhesion Molecules/blood , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Flow Cytometry , Fluorescent Antibody Technique , Humans , Microfilament Proteins/blood , Microscopy, Confocal , Phosphoproteins/blood , Phosphorylation , Platelet Function Tests , Protein Multimerization , Regional Blood Flow , Signal Transduction , Structure-Activity Relationship , Time Factors
7.
Platelets ; 22(3): 217-27, 2011.
Article in English | MEDLINE | ID: mdl-21244222

ABSTRACT

Platelet activation is strongly affected by nitric oxide/cyclic GMP (NO/cGMP) signaling involving cGMP-dependent protein kinase I (cGKI). Previously it was shown that interaction of the cGKI substrate IRAG with InsP(3)RI is essential for NO/cguanosine monophosphate (GMP)-dependent inhibition of platelet aggregation in vitro and in vivo. However, the role of Inositol-trisphosphate receptor associated cGMP kinase substrate (IRAG) for platelet adhesion or granule secretion was unknown. Here, we analysed the functional role of IRAG for platelet activation. Murine IRAG-deficient platelets displayed enhanced aggregability towards several agonists (collagen, thrombin and TxA2). NO- or cGMP-dependent inhibition of agonist induced ATP- or 5-HT secretion from dense granules, and P-selectin secretion from alpha granules was severely affected in IRAG-deficient platelets. Concomitantly, the effect of NO/cGMP on platelet aggregation was strongly reduced in IRAG-deficient platelets. Furthermore, GPIIb/IIIa-mediated adhesion of platelets to fibrinogen could only weakly be inhibited in IRAG-deficient mice contrary to wild-type (WT) mice. Our results suggest that signaling via IRAG is essential for NO/cGMP-dependent inhibition of platelet activation regarding granule secretion, aggregation and adhesion. This platelet disorder might cause that the bleeding time of IRAG-deficient mice was reduced.


Subject(s)
Blood Platelets/metabolism , Cyclic GMP/blood , Membrane Proteins/blood , Nitric Oxide/blood , Phosphoproteins/blood , Platelet Activation/physiology , Animals , Blood Platelets/enzymology , Cyclic GMP-Dependent Protein Kinases/blood , Enzyme Activators/pharmacology , Humans , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Signal Transduction
8.
Thromb Res ; 127(1): e8-14, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056902

ABSTRACT

In this study the effects of nitric oxide (NO) donors on intracellular free calcium ([Ca(2+)](i)) in human platelets was examined. Inhibition of guanylyl cyclase (GC) with either methylene blue or ODQ slightly inhibited the ability of submaximal concentrations of thrombin to increase [Ca(2+)](i) which suggests that a small portion of the thrombin mediated increase in [Ca(2+)](i) was due to an increase in NO and subsequent increase in cGMP and activation of cGMP dependent protein kinase (cGPK). Thrombin predominantly increases [Ca(2+)](i) by stimulating store-operated Ca(2+) entry (SOCE). The NO donor GEA3162 was previously shown to stimulate SOCE in some cells. In platelets GEA3162 had no effect to increase [Ca(2+)](i) however it inhibited the ability of thrombin to increase [Ca(2+)](i) and this effect was reversed by ODQ. The addition of low concentrations (2.0 - 20 nM) of the NO donor sodium nitroprusside (SNP) slightly potentiated the ability of thrombin to increase [Ca(2+)](i) whereas higher concentrations (>200 nM) of SNP inhibited thrombin induced increases in [Ca(2+)](i). Both of these effects of SNP were reversed by ODQ which implies that they were both mediated by cGPK. Ba(2+) influx was stimulated by low concentrations (2.0 nM) of SNP and inhibited by high concentrations (>200 nM) of SNP and both effects were inhibited by ODQ. Previous studies showed that Ba(2+) influx was blocked by the SOCE inhibitors 2-aminoethoxydipheny borate and diethylstilbestrol. It was concluded that low levels of SNP can stimulate SOCE in platelets and this effect may account for the increased aggregation and secretion previously observed with low concentrations of NO donors. Of the proteins known to be involved in SOCE (e.g. stromal interaction molecule 1 (Stim1), Stim2 and Orai1) only Stim2 has cGPK phosphorylation sites. The possibility that Stim2 phosphorylation regulates SOCE in platelets is discussed.


Subject(s)
Blood Platelets/drug effects , Calcium/blood , Nitric Oxide Donors/pharmacology , Nitric Oxide/blood , Nitroprusside/pharmacology , Triazoles/pharmacology , Blood Platelets/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cell Adhesion Molecules/blood , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/blood , Humans , Membrane Proteins/blood , Neoplasm Proteins/blood , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/blood , Phosphorylation , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , Thrombin/metabolism , Time Factors
9.
Platelets ; 20(7): 478-86, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19852686

ABSTRACT

Nitric oxide (NO)-mediated inhibition of platelet function occurs primarily through elevations in cGMP, although cGMP-independent mechanisms such as S-nitrosylation have been suggested as alternative NO-signaling pathways. In the present study we investigated the potential for S-nitrosylation to act as a NO-mediated cGMP-independent signaling mechanism in platelets. The NO-donor, S-nitrosoglutathione (GSNO), induced a concentration-dependent inhibition of platelet adhesion to immobilized collagen. In the presence of the soluble guanylyl cyclase inhibitor, ODQ, NO-mediated activation of the cGMP/protein kinase G signaling pathway was ablated. However, ODQ failed to completely abolish the inhibitory effect of NO on collagen-mediated adhesion, confirming that cGMP-independent signaling events contribute to the regulation of platelet adhesion by NO. Biotin-switch analysis of platelets demonstrated the presence of several S-nitrosylated proteins under basal conditions. Treatment of platelets with exogenous NO-donors, at concentrations that inhibited platelet adhesion, increased the number of S-nitrosylated bands and led to hyper-nitrosylation of basally S-nitrosylated proteins. The extent of S-nitrosylation in response to exogenous NO was unaffected by platelet activation. Importantly, platelet activation in the absence of exogenous NO failed to increase S-nitrosylation beyond basal levels, indicating that platelet-derived NO was unable to induce this type of protein modification. Our data demonstrate that S-nitrosylation of platelet proteins in response to exogenous NO may act as a potentially important cGMP-independent signaling mechanism for controlling platelet adhesion.


Subject(s)
Collagen/blood , Cyclic GMP/blood , Nitric Oxide/pharmacology , Platelet Adhesiveness/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Proteins/metabolism , Cyclic GMP-Dependent Protein Kinases/blood , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Humans , Immunoblotting , Nitric Oxide/blood , Nitric Oxide Donors/blood , Nitric Oxide Donors/pharmacology , Oxadiazoles/pharmacology , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Function Tests , Quinoxalines/pharmacology , S-Nitrosoglutathione/pharmacology , Signal Transduction/drug effects
10.
Handb Exp Pharmacol ; (191): 533-48, 2009.
Article in English | MEDLINE | ID: mdl-19089344

ABSTRACT

Platelets are specialized adhesive cells that play a key role in normal and pathological hemostasis through their ability to rapidly adhere to subendothelial matrix proteins (platelet adhesion) and to other activated platelets (platelet aggregation). NO plays a crucial role in preventing platelet adhesion and aggregation. In platelets, cGMP synthesis is catalyzed by sGC, whereas PDE2, PDE3 and PDE5 are responsible for cGMP degradation. Stimulation of cGK by cGMP leads to phosphorylation of multiple target substrates. These substrates inhibit elevation of intracellular calcium, integrin activation, cytoskeletal reorganization, and platelet granule secretion, events normally associated with platelet activation. The NO/cGMP pathway also plays a significant role in many other blood cell types in addition to platelets. In leukocytes, depending on the specific cell type, cGMP signaling regulates gene expression, differentiation, migration, cytokine production, and apoptosis.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/blood , Cyclic GMP/blood , Signal Transduction , Animals , Blood Platelets/metabolism , Cyclic GMP/biosynthesis , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Humans , Leukocytes/metabolism , Nitric Oxide/metabolism , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology
11.
Blood ; 103(7): 2593-600, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14644996

ABSTRACT

Platelets play a key role in hemostasis through their ability to rapidly adhere to activated or injured endothelium, subendothelial matrix proteins, and other activated platelets. A strong equilibrium between activating and inhibiting processes is essential for normal platelet and vascular function, impairment of this equilibrium being associated with either thrombophilic or bleeding disorders. Both cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) have been established as crucial and synergistic intracellular messengers that mediate the effects of platelet inhibitors such as nitric oxide (NO) and prostacyclin (PG-I2). However, it was recently suggested that a rapid cGMP/cGMP-dependent protein kinase (cGK)-mediated extracellular signal-related kinase (ERK) phosphorylation promotes platelet activation. This hypothesis was examined here by evaluating established and proposed cGK activators/inhibitors with respect to their capacity to promote either platelet activation or inhibition. In particular, the regulatory role of cGK for ERK phosphorylation and thrombin-, thromboxane-, and VWF-induced platelet activation was investigated. The data obtained do not support the concept that cGK-mediated ERK phosphorylation promotes platelet activation but confirm the inhibitory role of cGK in platelet function. One explanation for these discrepancies is the novel finding that extracellular cGMP analogs potently and rapidly inhibit thrombin-, thromboxane-, and VWF-induced human platelet signaling and activation by a cGK-independent mechanism.


Subject(s)
Blood Platelets/physiology , Cyclic GMP-Dependent Protein Kinases/blood , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Blood Platelets/drug effects , Blood Platelets/enzymology , Calcium/blood , Cyclic GMP/blood , Humans , Kinetics , Nitric Oxide Donors/pharmacology , Platelet Aggregation/drug effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , von Willebrand Factor/pharmacology
12.
Blood ; 103(7): 2601-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14684423

ABSTRACT

Glycoprotein Ib-IX-V (GPIb-IX-V) mediates platelet tethering to von Willebrand factor (VWF), recruiting platelets into the thrombus, and activates integrin alphaIIbbeta3 through a pathway that is dependent on Src kinases. In addition, recent reports indicate that activation of alphaIIbbeta3 by VWF is dependent on protein kinase G (PKG) and mitogen-activated protein (MAP) kinases. The present study compares the importance of these signaling pathways in the activation of alphaIIbbeta3 by GPIb-IX-V. In contrast to a recent report, VWF did not promote an increase in cyclic guanosine monophosphate (cGMP), while agents that elevate cGMP, such as the nitrous oxide (NO) donor glyco-SNAP-1 (N-(beta-D-glucopyranosyl)-N2-acetyl-S-nitroso-D,L-penicillaminamide) or the type 5 phosphosdiesterase inhibitor, sildenafil, inhibited rather than promoted activation of alphaIIbbeta3 by GPIb-IX-V and blocked aggregate formation on collagen at an intermediate rate of shear (800 s(-1)). Additionally, sildenafil increased blood flow in a rabbit model of thrombus formation in vivo. A novel inhibitor of the MAP kinase pathway, which is active in plasma, PD184161, had no effect on aggregate formation on collagen under flow conditions, whereas a novel inhibitor of Src kinases, which is also active in plasma, PD173952, blocked this response. These results demonstrate a critical role for Src kinases but not MAP kinases in VWF-dependent platelet activation and demonstrate an inhibitory role for cGMP-elevating agents in regulating this process.


Subject(s)
Blood Platelets/physiology , Cyclic GMP-Dependent Protein Kinases/blood , Mitogen-Activated Protein Kinases/blood , Platelet Activation/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins , Animals , Blood Platelets/drug effects , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Humans , Kinetics , Mice , Mice, Knockout , Nitric Oxide Donors/pharmacology , Receptors, Antigen, B-Cell/blood , von Willebrand Factor/pharmacology
13.
Nitric Oxide ; 5(3): 233-42, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11384196

ABSTRACT

In previous studies, a strong synergism between low concentrations of hydrogen peroxide and nitric oxide in the inhibition of agonist-induced platelet aggregation has been established and may be due to enhanced formation of cyclic GMP. In this investigation, hydrogen peroxide and NO had no effect on the activity of pure soluble guanylyl cyclase or its activity in platelet lysates and cytosol. H(2)O(2) was found to increase the phosphorylation of vasodilator-stimulated phosphoprotein (VASP), increasing the amount of the 50-kDa form that results from phosphorylation at serine(157). This occurs both in the presence and in the absence of low concentrations of NO, even at submicromolar concentrations of the peroxide, which alone was not inhibitory to platelets. These actions of H(2)O(2) were inhibited to a large extent by an inhibitor of cyclic AMP-dependent protein kinase, even though H(2)O(2) did not increase cyclic AMP. This inhibitor reversed the inhibition of platelets induced by combinations of NO and H(2)O(2) at low concentrations. The results suggest that the action on VASP may be one site of action of H(2)O(2) but that this event alone does not lead to inhibition of platelets; another unspecified action of NO is required to complete the events required for inhibition.


Subject(s)
Blood Platelets/physiology , Carbazoles , Cell Adhesion Molecules/metabolism , Cyclic GMP/blood , Guanylate Cyclase/blood , Hydrogen Peroxide/pharmacology , Indoles , Nitric Oxide/pharmacology , Phosphoproteins/metabolism , Platelet Aggregation/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Alkaloids/pharmacology , Blood Platelets/drug effects , Blood Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/blood , Cytosol/enzymology , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Hydrazines/pharmacology , In Vitro Techniques , Indazoles/pharmacology , Kinetics , Microfilament Proteins , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitrogen Oxides , Phosphoserine/blood , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/pharmacology , Subcellular Fractions/metabolism , Thrombin/pharmacology
14.
Mol Biol Cell ; 9(10): 2933-47, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9763453

ABSTRACT

In neutrophils activated to secrete with formyl-methionyl-leucyl-phenylalanine, intermediate filaments are phosphorylated transiently by cyclic guanosine monophosphate (cGMP)-dependent protein kinase (G-kinase). cGMP regulation of vimentin organization was investigated. During granule secretion, cGMP levels were elevated and intermediate filaments were transiently assembled at the pericortex to areas devoid of granules and microfilaments. Microtubule and microfilament inhibitors affected intermediate filament organization, granule secretion, and cGMP levels. Cytochalasin D and nocodazole caused intermediate filaments to assemble at the nucleus, rather than at the pericortex. cGMP levels were elevated in neutrophils by both inhibitors; however, with cytochalasin D, cGMP was elevated earlier and granule secretion was excessive. Nocodazole did not affect normal cGMP elevations, but specific granule secretion was delayed. LY83583, a guanylyl cyclase antagonist, inhibited granule secretion and intermediate filament organization, but not microtubule or microfilament organization. Intermediate filament assembly at the pericortex and secretion were partially restored by 8-bromo-cGMP in LY83583-treated neutrophils, suggesting that cGMP regulates these functions. G-kinase directly induced intermediate filament assembly in situ, and protein phosphatase 1 disassembled filaments. However, in intact cells stimulated with formyl-methionyl-leucyl-phenylalanine, intermediate filament assembly is focal and transient, suggesting that vimentin phosphorylation is compartmentalized. We propose that, in addition to changes in microfilament and microtubule organization, granule secretion is also accompanied by changes in intermediate filament organization, and that cGMP regulates vimentin filament organization via activation of G-kinase.


Subject(s)
Cyclic GMP/blood , Cytoplasmic Granules/physiology , Intermediate Filaments/ultrastructure , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/physiology , Neutrophils/ultrastructure , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Aminoquinolines/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/blood , Cytochalasin D/pharmacology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/ultrastructure , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Intermediate Filaments/drug effects , Intermediate Filaments/physiology , Kinetics , Microtubules/drug effects , Microtubules/physiology , Microtubules/ultrastructure , Models, Biological , Neutrophils/drug effects , Nocodazole/pharmacology , Phosphorylation , Signal Transduction , Vimentin/blood
15.
J Leukoc Biol ; 60(1): 94-100, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8699130

ABSTRACT

We studied the effect of exogenous nitric oxide (NO) on migration of rabbit peritoneal neutrophils. Exogenous NO enhanced random migration of neutrophils in a concentration-dependent way. An optimally stimulatory effect was observed with 0.5 microM NO, whereas at higher NO concentrations the enhancing effect decreased again. NO caused a rapid and transient increase in intracellular guanosine-3',5'-cyclic monophosphate (cGMP) levels. The enhancing effect of NO on random migration was largely reversed by the inhibitors of cGMP accumulation, LY-83583 and methylene blue, and by the antagonists of cGMP-dependent protein kinase, 8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-8-Br-cGMPS) and 8-(4-chlorophenylthio)-guanosine-3',5'-cyclic monophosphorothioate (Rp-8-pCPT-cGMPS). These observations strongly suggest that the enhancement of random migration by NO is mediated by cGMP and cGMP-dependent protein kinase. The effect of NO on migration did not occur in the absence of extracellular calcium. Although NO did not induce a measurable elevation of intracellular free calcium, pre-incubation with the intracellular calcium chelator Fura-2/AM abolished the enhancing effect of NO. It appears therefore that a small change in the level of cytoplasmic free calcium does play a role in the enhancement of random migration by NO. High concentrations of NO were found to inhibit chemotaxis induced by an optimal concentration of the chemotactic peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP). This inhibitory effect was also dependent on the presence of extracellular calcium. A role for cGMP in the inhibition of fMLP-induced chemotaxis by NO is not supported by our measurements of intracellular cGMP levels. In contrast to the effects on fMLP, NO did not affect chemotaxis induced by the phorbol ester PMA. In conclusion, we show that NO, not derived from NO donors but applied directly, may stimulate or inhibit neutrophil migration, dependent on the concentration. The enhancing effect of NO on random migration is mediated by cGMP, which emphasizes the importance of this second messenger as a modulator of neutrophil functional.


Subject(s)
Chemotaxis, Leukocyte/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/physiology , Nitric Oxide/pharmacology , Aminoquinolines/pharmacology , Animals , Calcium/blood , Calcium/pharmacology , Chemotaxis, Leukocyte/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/blood , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Kinetics , Magnesium/pharmacology , Neutrophils/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Rabbits , Tetradecanoylphorbol Acetate/pharmacology
16.
Eur J Biochem ; 223(2): 543-51, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8055924

ABSTRACT

The Ca2+ responses of rat platelets are dominated by the influx of extracellular Ca2+ across the plasma membrane [Heemskerk, J. W. M., Feijge, M. A. H., Rietman, E. & Hornstra, G. (1991) FEBS Lett. 284, 223], which allows the study of Ca2+ entry into these cells by measuring increases in cytosolic Ca2+ concentration, [Ca2+]i. Several pieces of evidence indicated that, as in human platelets [Sage, S. O., Reast, R., & Rink, T. J. (1990) Biochem. J. 265, 675-680; Alonso, M., Alvarez, J., Montero, M., Sanchez, A. & García-Sancho, J. (1991) Biochem. J. 280, 783-789], agonist-stimulated Ca2+ entry was linked to the mobilisation of Ca2+ from intracellular stores: there was good correlation between the potency of receptor agonists in elevating [Ca2+]i in the presence or absence of external CaCl2; agonist-induced Ca2+ entry was inhibited to a similar degree as internal mobilisation by activators of cAMP-dependent or cGMP-dependent protein kinase or by the phospholipase C inhibitor, U73122; thapsigargin (an inhibitor of endomembrane Ca(2+)-ATPases) evoked store depletion and Ca2+ entry, which were both reduced by prior activation of cAMP-dependent or cGMP-dependent protein kinase but were not affected by U73122. In platelets with depleted Ca2+ stores, the addition of CaCl2 resulted in a considerable entry of Ca2+ which was insensitive to cAMP-dependent and cGMP-dependent protein kinase activation. In control platelets with full Ca2+ stores, CaCl2 potentiated the thrombin-induced generation of myo-inositol phosphates, suggesting that Ca2+ entry potentiated phospholipase C activity. Taken together, these results indicate that Ca2+ entry in rat platelets, (a) is mostly secondary to store depletion, (b) is not directly downregulated by cAMP-dependent and cGMP-dependent protein kinase, but indirectly by inhibition of store depletion, (c) can proceed in the absence of phospholipase C activation, but is stimulated by this activity probably by increased mobilisation of Ca2+ from the stores. These results lead to the concept that a major part of receptor-mediated Ca2+ entry in rat platelets is regulated in an indirect way by factors that stimulate or inhibit the degree of Ca2+ mobilisation from the internal stores.


Subject(s)
Blood Platelets/metabolism , Calcium/blood , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic GMP-Dependent Protein Kinases/blood , Type C Phospholipases/blood , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Blood Platelets/drug effects , Calcium Chloride/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Drug Synergism , Egtazic Acid/pharmacology , Estrenes/pharmacology , Male , Platelet Activation/drug effects , Prostaglandin Endoperoxides, Synthetic/pharmacology , Pyrrolidinones/pharmacology , Rats , Rats, Wistar , Terpenes/pharmacology , Thapsigargin , Thrombin/pharmacology , Thromboxane A2/analogs & derivatives , Thromboxane A2/pharmacology , Type C Phospholipases/antagonists & inhibitors
17.
J Biol Chem ; 269(20): 14509-17, 1994 May 20.
Article in English | MEDLINE | ID: mdl-8182057

ABSTRACT

The vasodilator-stimulated phosphoprotein (VASP) is a major substrate for cAMP-dependent- (cAK) and cGMP-dependent protein kinase (cGK) in human platelets and other cardiovascular cells. To identify the VASP phosphorylation sites, purified VASP was phosphorylated by either protein kinase and subjected to trypsin, V8 and Lys-C proteolysis. The phosphorylated proteolytic fragments obtained were separated by reversed phase high performance liquid chromatography. Sequence analysis of the phosphorylated peptides and 32P measurement of the released 32P-labeled amino acids revealed three phosphorylation sites: a serine 1-containing site (LRKVSKQEEA), a serine 2-containing site (HIERRVSNAG), and a threonine-containing site (MNAVLARRRKATQVGE). Additional experiments with purified VASP demonstrated that both cAK and cGK phosphorylated serine 2 rapidly and the threonine residue slowly, whereas cGK phosphorylated the serine 1 residue more rapidly than the cAK. These differences in the phosphorylation rates of VASP by the two protein kinases were also observed with synthetic peptides corresponding to the sequences of the three identified phosphorylation sites. These experiments also established the synthetic peptide serine 1 as one of the best in vitro cGK substrates and the serine 2-containing site as the site responsible for the phosphorylation-induced mobility shift of VASP in sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Experiments with 32P-labeled platelets provided evidence that VASP is phosphorylated at the same three identified sites also in intact cells and that selective activation of cAK or cGK primarily increased the phosphorylation of both serine 2 and serine 1 but not threonine. Our results demonstrated overlapping substrate specificities of cAK and cGK in vitro and in intact cells. However, important quantitative and qualitative differences between cAK- and cGK-mediated phosphorylation of the focal adhesion protein VASP in human platelets were also observed, suggesting distinct functions of the two types of cyclic nucleotide-mediated VASP phosphorylation.


Subject(s)
Blood Platelets/enzymology , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic GMP-Dependent Protein Kinases/blood , Microfilament Proteins/blood , Phosphoproteins/blood , Amino Acid Sequence , Autoradiography , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Microfilament Proteins/chemistry , Microfilament Proteins/isolation & purification , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphorus Radioisotopes , Phosphorylation , Phosphoserine/analysis , Phosphothreonine/analysis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL