4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: not found
      • Article: not found

      Reduced Vascular NO Bioavailability in Diabetes Increases Platelet Activation In Vivo

      Read this article at

      ScienceOpenPublisherPubMed
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Platelet activation is a feature of cardiovascular disease that is also characterized by endothelial dysfunction. The direct relationship between impaired endothelium-derived NO bioavailability and platelet activation remains unclear. We investigated whether acute inhibition of NO production modulates platelet activation in mice and whether specific rescue of endothelial function in diabetes modifies platelet activation. Intravenous injection of the NO synthase inhibitor N(G)-nitro-L-arginine methyl ester in wild-type (WT) mice significantly reduced platelet vasodilator-stimulated phosphoprotein (VASP) phosphorylation and increased platelet surface expression of P-selectin, CD40 ligand, and fibrinogen platelet binding, demonstrating that NO production exerts tonic inhibition of platelet activation in mice. Diabetes was induced by streptozotocin injection in WT or endothelial-targeted guanosine 5'-triphosphate cyclohydrolase I (GCH)-transgenic (GCH-Tg) mice protected from endothelial dysfunction in diabetes by sustained levels of tetrahydrobiopterin in vascular endothelium. Platelet VASP phosphorylation was significantly reduced in diabetic WT but not in diabetic GCH-Tg mice. P-selectin, CD40 ligand expression, and fibrinogen binding were increased in diabetic WT mice but remained unchanged compared with controls in endothelial-targeted GCH-Tg mice. Platelet activation results from acute and chronic reduction in NO bioactivity. Rescue of platelet activation in diabetes by endothelial-specific restoration of NO production demonstrates that platelet function in vivo is principally regulated by endothelium-derived NO. Endothelial dysfunction caused by uncoupling of endothelial NO synthase is well described in diabetes mellitus and may lead to platelet activation. Acute loss of systemic NO bioavailability causes platelet activation. eNOS uncoupling prevention in diabetes preserved systemic NO bioavailability and maintained a physiological platelet state without activation in vivo.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: not found

          CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells.

          CD40 ligand (CD40L, CD154), a transmembrane protein structurally related to the cytokine TNF-alpha, was originally identified on stimulated CD4+ T cells, and later on stimulated mast cells and basophils. Interaction of CD40L on T cells with CD40 on B cells is of paramount importance for the development and function of the humoral immune system. CD40 is not only constitutively present on B cells, but it is also found on monocytes, macrophages and endothelial cells, suggesting that CD40L has a broader function in vivo. We now report that platelets express CD40L within seconds of activation in vitro and in the process of thrombus formation in vivo. Like TNF-alpha and interleukin-1, CD40L on platelets induces endothelial cells to secrete chemokines and to express adhesion molecules, thereby generating signals for the recruitment and extravasation of leukocytes at the site of injury. Our results indicate that platelets are not only involved in haemostasis but that they also directly initiate an inflammatory response of the vessel wall.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Tetrahydrobiopterin-dependent preservation of nitric oxide-mediated endothelial function in diabetes by targeted transgenic GTP-cyclohydrolase I overexpression.

            Increased production of reactive oxygen species and loss of endothelial NO bioactivity are key features of vascular disease states such as diabetes mellitus. Tetrahydrobiopterin (BH4) is a required cofactor for eNOS activity; pharmacologic studies suggest that BH4 may mediate some of the adverse effects of diabetes on eNOS function. We have now investigated the importance and mechanisms of BH4 availability in vivo using a novel transgenic mouse model with endothelial-targeted overexpression of the rate-limiting enzyme in BH4 synthesis, guanosine triphosphate-cyclohydrolase I (GTPCH). Transgenic (GCH-Tg) mice demonstrated selective augmentation of endothelial BH4 levels. In WT mice, induction of diabetes with streptozotocin (STZ) increased vascular oxidative stress, resulting in oxidative loss of BH4, forming BH2 and biopterin. Endothelial cell superoxide production in diabetes was increased, and NO-mediated endothelium-dependent vasodilatation was impaired. In diabetic GCH-Tg mice, superoxide production from the endothelium was markedly reduced compared with that of WT mice, endothelial BH4 levels were maintained despite some oxidative loss of BH4, and NO-mediated vasodilatation was preserved. These findings indicate that BH4 is an important mediator of eNOS regulation in diabetes and is a rational therapeutic target to restore NO-mediated endothelial function in diabetes and other vascular disease states.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Analysis and regulation of vasodilator-stimulated phosphoprotein serine 239 phosphorylation in vitro and in intact cells using a phosphospecific monoclonal antibody.

              The development and functional analysis of a monoclonal antibody (16C2) are reported; the antibody recognizes vasodilator-stimulated phosphoprotein (VASP; an established substrate of both cAMP- and cGMP-dependent protein kinase) only when serine 239 is phosphorylated. VASP serine 239 represents one of the best characterized cGMP-dependent protein kinase phosphorylation sites in vitro and in intact cells. Experiments with purified, recombinant human VASP and various VASP constructs with mutated phosphorylation sites (S157A, S239A, T278A) and experiments with intact cells (human/rat platelets and other cells) treated with cyclic nucleotide-elevating agents demonstrated the specificity of the monoclonal antibody 16C2. Quantitative analysis of the VASP shift from 46 to 50 kDa (indicating VASP serine 157 phosphorylation) and the appearance of VASP detected by the 16C2 monoclonal antibody (VASP serine 239 phosphorylation) in human platelets stimulated by selective protein kinase activators confirmed that serine 239 is the VASP phosphorylation site preferred by cGMP-dependent protein kinase in intact cells. Immunofluorescence experiments with human platelets treated with cGMP analogs showed that the 16C2 monoclonal antibody also detects VASP serine 239 phosphorylation in situ at established intracellular localization sites. Analysis of VASP serine 239 phosphorylation by the 16C2 antibody appears to be the best method presently available to measure cGMP-dependent protein kinase activation in intact cells. Also, the 16C2 antibody promises to be an excellent tool for the evaluation of VASP function in intact cells.
                Bookmark

                Author and article information

                Journal
                Arteriosclerosis, Thrombosis, and Vascular Biology
                ATVB
                Ovid Technologies (Wolters Kluwer Health)
                1079-5642
                1524-4636
                September 2004
                September 2004
                : 24
                : 9
                : 1720-1726
                Affiliations
                [1 ]From the Department of Cardiovascular Medicine (A.S., N.J.A., S.C., C.A.L., S.N., K.M.C.), University of Oxford, United Kingdom; and Medizinische Klinik (A.S., J.B.), Institut für Klinische Biochemie und Pathobiochemie (M.E.), Julius-Maximilians-Universität Würzburg, Germany.
                Article
                10.1161/01.ATV.0000138072.76902.dd
                15242858
                66463791-c1a0-42d6-becc-aca3ac3490b0
                © 2004
                History

                Comments

                Comment on this article