Platelet Aggregation Inhibiting and Anticoagulant Effects of Oligoamines, XXVII Inhibition of Leucocyte Adherence to Endothelium by the Oligoamine RE 1492C and the NO-Donor RE 2047

1995 ◽  
Vol 328 (2) ◽  
pp. 125-126 ◽  
Author(s):  
Klaus Rehse ◽  
Thomas Ciborski ◽  
Bernd Müller
Amino Acids ◽  
2021 ◽  
Author(s):  
Dimitrios Tsikas

AbstractNitrosylation of sulfhydryl (SH) groups of cysteine (Cys) moieties is an important post-translational modification (PTM), often on a par with phosphorylation. S-Nitrosoalbumin (ALB-Cys34SNO; SNALB) in plasma and S-nitrosohemoglobin (Hb-Cysβ93SNO; HbSNO) in red blood cells are considered the most abundant high-molecular-mass pools of nitric oxide (NO) bioactivity in the human circulation. SNALB per se is not an NO donor. Yet, it acts as a vasodilator and an inhibitor of platelet aggregation. SNALB can be formed by nitrosation of the sole reduced Cys group of albumin (Cys34) by nitrosating species such as nitrous acid (HONO) and nitrous anhydride (N2O3), two unstable intermediates of NO autoxidation. SNALB can also be formed by the transfer (S-transnitrosylation) of the nitrosyl group (NO+) of a low-molecular-mass (LMM) S-nitrosothiol (RSNO) to ALB-Cys34SH. In the present study, the effects of LMM thiols on the inhibitory potential of ALB-Cys34SNO on human washed platelets were investigated. ALB-Cys34SNO was prepared by reacting n-butylnitrite with albumin after selective extraction from plasma of a healthy donor on HiTrapBlue Sepharose cartridges. ALB-Cys34SNO was used in platelet aggregation measurements after extended purification on HiTrapBlue Sepharose and enrichment by ultrafiltration (cutoff, 20 kDa). All tested LMM cysteinyl thiols (R-CysSH) including l-cysteine and L-homocysteine (at 10 µM) were found to mediate the collagen-induced (1 µg/mL) aggregation of human washed platelets by SNALB (range, 0–10 µM) by cGMP-dependent and cGMP-independent mechanisms. The LMM thiols themselves did not affect platelet aggregation. It is assumed that the underlying mechanism involves S-transnitrosylation of SH groups of the platelet surface by LMM RSNO formed through the reaction of SNALB with the thiols: ALB-Cys34SNO + R-CysSH ↔ ALB-Cys34SH + R-CysSNO. Such S-transnitrosylation reactions may be accompanied by release of NO finally resulting in cGMP-dependent and cGMP-independent mechanisms.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4971-4971
Author(s):  
Yifeng WU ◽  
Alan D Michelson ◽  
Andrew L Frelinger

Abstract Introduction Nitric oxide (NO) released by endothelial cells interacts with platelets in which it stimulates soluble guanylate cyclase (sGC), thereby increasing platelet cyclic guanosine monophosphate (cGMP) and inhibiting platelet activation. Stimulation of sGC in other cells has been suggested as an attractive target for intervention in a range of diseases including pulmonary arterial hypertension, heart failure, and diabetes mellitus. Riociguat, the first FDA-approved sGC stimulator, potently increases platelet cGMP and inhibits platelet aggregation in washed platelets. Because riociguat binds to plasma proteins, higher concentrations of riociguat are required to inhibit platelet function in whole blood. However the potential synergistic inhibition of platelet function by riociguat and NO has not been well studied. Goal To investigate the possible synergistic effects of riociguat and NO on platelet inhibition and to determine the effects of protein binding. Methods Platelet-rich plasma (PRP) was prepared from citrate (3.2%) anticoagulated whole blood collected from healthy donors following informed consent. Riociguat (10 mM) in DMSO and DETA-NONOate 10 mM (an NO donor) in 10 mM NaOH were stored at -80°C until use. PRP was diluted 10-fold in either HEPES-Tyrode's buffer or platelet poor plasma (PPP), then incubated with vehicle or riociguat 1, 10, or 100 µM, alone or in combination with DETA-NONOate 16, 31, or 250 µM for 30 minutes, then analyzed by flow cytometry. Platelet surface activated GPIIb-IIIa (detected by monoclonal antibody PAC1) and platelet surface P-selectin were measured with and without activation by ADP 5 µM or thrombin receptor activating peptide (TRAP) 5 µM. For light transmission platelet aggregation (LTA) and 96-well platelet aggregation, PRP was used without dilution. Results For PRP diluted in buffer, riociguat and DETA-NONOate each produced concentration-dependent inhibition of ADP- and TRAP-stimulated platelet activation, as reported by platelet surface activated GPIIb-IIIa (Figure A) and P-selectin, and a synergistic inhibitory effect was observed when riociguat and DETA-NONOate were combined (for platelet surface activated GPIIb-IIIa, 40% inhibition with 1 µM riociguat alone; 30% inhibition with 31 µM DETA-NONOate alone; 90% inhibition with 1 µM riociguat and 31 µM DETA-NONOate combined). In contrast, when PRP was diluted in PPP, the concentrations of riociguat alone and DETA-NONOate alone needed to inhibit activation were dramatically increased and the combination of 1 µM riociguat and 31 µM DETA-NONOate produced less than 10% inhibition of platelet surface activated GPIIb-IIIa (Figure B). Synergistic inhibition in plasma was observed when DETA-NONOate was increased to 250 µM. Based on these results, a sub-threshold concentration of DETA-NONOate was chosen for investigation of the effects of riociguat on platelet aggregation. Using ADP 5 µM, TRAP, 2 µM, or collagen 2 µg/mL, riociguat alone at 10 µM (Figure C) and DETA-NONOate alone at 31 µM showed no inhibition of platelet aggregation. However, in the presence of 31 µM DETA-NONOate, riociguat showed a concentration-dependent inhibition of aggregation by each agonist (Figure C). Conclusions Platelets exposed to riociguat in combination with sub-threshold concentrations of NO, such as may occur in microvessels adjacent to the endothelial layer, are inhibited from undergoing platelet activation and aggregation. The presence of plasma proteins blunts the effects of riociguat and has even larger effects on the NO donor, DETA-NONOate. Taken together, these data suggest that NO potently synergizes with riociguat to inhibit platelet activation and aggregation, but in vivo this effect likely only occurs immediately adjacent to endothelial cells where NO concentrations are highest. Figure. Figure. Disclosures Michelson: Alnylam, Instrumentation Laboratory, Surface Oncology: Consultancy; AstraZeneca, Chiesi, Dova, Janssen, LightIntegra, Megakaryon, Remora: Other: Scientific Advisory Board; Baxalta, Cellular Preservation Technologies, Ionis, Ironwood, Medtronic, Megakaryon, Pfizer, Sysmex: Research Funding. Frelinger:Surface Oncology: Consultancy; Cellular Preservation Technologies, Ironwood, Ionis, Medtronic, Megakaryon, Pfizer and Sysmex: Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3364-3364
Author(s):  
Jean Leandro Santos ◽  
Ednir Vizioli de Oliveira ◽  
Maria Elisa Lopes Pires ◽  
Thais Regina Ferreira de Melo ◽  
Fernando Ferreira Costa ◽  
...  

Abstract Abstract 3364 Introduction. Platelets are involved in atherothrombosis as a consequence of their inappropriate and sustained activation, being thus considered an important target for pharmacological interventions. Aspirin (ASA) is the current drug standard of care of most patients; however the therapy is limited and presents several inconveniences such as lack of effectiveness and bleeding. The use of hybridization strategy allows obtaining compounds with pharmacokinetic and pharmacodynamic properties improved. In this work, we describe new compounds with significant antiplatelet activity induced by ADP. These compounds 2-(2-((4-methyl-1,2,5-oxadiazole-2-oxide)methylene)hydrazinecarbonyl) phenyl acetate (Lapdesf 1) and 2-(2-((4-phenyl-1,2,5-oxadiazole-2-oxide)methylene)hydrazinecarbonyl) phenyl acetate (Lapdesf 2) were structurally planned by modification of acetyl salicylic acid (ASA) based on the use of acylhydrazone subunit, previously reported to contribute to inhibit platelet aggregation. Furthermore, 1,2,5-oxadiazole N-oxide subunit known by its NO-donor properties were included in the hybrid structure in order to contribute for the antiplatelet activity. Methods. 1. Detection of nitrite. A solution of the appropriate compound (20 μL) in DMSO was added to 2 mL of a mixture of 50 mM phosphate buffer (pH 7.4) and methanol (1:1, v:v), containing 5 mM of L-cysteine. The final concentration of the compound was 10–4M. After 1 h at 37 °C, 1 mL of the reaction mixture was treated with 250 μL of Griess reagent. After 10 min at room temperature, the absorbance was measured at 540 nm using a spectrophotometer. Standard sodium nitrite solutions (10–80 nmol/mL) were used to construct the calibration curve. The yields of nitrite are expressed as % NO2– (mol/mol). 2. Antiplatelet activity. Blood was withdrawn from mice central artery and mixed with 3.8% trisodium citrate (9:1 v/v). Platelet-rich plasma (PRP) was prepared by centrifugation at 375 × g for 10 min at room temperature. The platelet-poor plasma (PPP) was prepared by centrifugation of the pellet at 1800 × g for 10 min at room temperature. Platelet aggregation was monitored by the turbidimetric method of Born and Cross using aggregometer. PRP (300 μL) was incubated at 37 °C for 1 min with compounds before addition of the aggregating agent. Platelet aggregation was induced by ADP (10 μM). Compounds and ASA used as control were evaluated at 150 μM. 3. Tail vein bleeding time model. Mice were anesthetized (ketamine 75mg/kg; medetomidine 1 mg/kg IP). A transverse incision is made with a scalpel over a lateral tail vein at a position where the diameter of the tail is 2.25–2.5 mm. The tail was immersed in normal saline (37°C) in a hand-held test tube. This last one is rotated to prevent the shed blood from obscuring the incision site. The time from the incision to the cessation of bleeding is recorded. The results are expressed as mean time + SEM of 6 animals per group. The data were analyzed statistically with Student's t test at a significance level of P < 0.05. Results. 1. Detection of nitrite: Isosorbide dinitrate used as the control, induced 10.9 + 0.5 % of nitrite (mol/mol) formation while compound Lapdesf 1 and Lapdesf 2 induced 1.3 + 0.1 and 4.7 + 0.2, respectively. 2.Antiplatelet activity. The compounds inhibited platelet aggregation induced by ADP. The aggregation percentage in presence of Lapdesf 1 and Lapdesf 2 were 3.1 + 0,2 and 32 + 0,4 respectively. The aggregation percentage of control (ADP) and ASA were 40.3 + 0,3 and 57.5 + 0,4 respectively. 3. Tail vein bleeding time model. The control used showed an average of 52 + 15 seconds to stop bleeding while ASA demonstrated an average of 73 + 20 seconds. Lapdesf 1 and Lapdesf 2 showed an average of 113 + 25 seconds and 192 + 31 seconds. Conclusions. Results demonstrate that molecular hybridization approach used allows us to obtain new ASA derivatives with property of inhibited platelet aggregation induced by ADP. Lapdesf 1 and 2 demonstrated different NO-donor properties related to methyl and phenyl substitutions. Furthermore, both compounds prolonged tail vein bleeding time using mice model. These compounds could be new drug candidates, alternative to ASA, useful to prevent atherothrombosis events. Disclosures: No relevant conflicts of interest to declare.


1995 ◽  
Vol 73 (02) ◽  
pp. 297-303 ◽  
Author(s):  
Ruth Armstrong ◽  
Jane A May ◽  
Wolfgang Lösche ◽  
Stan Heptinstall

SummaryWhen whole blood is stirred there is a “spontaneous” platelet aggregation (SPA) which is presumed to be caused by proaggregatory factors released from platelets and other blood cells. Adding streptokinase (SK) to stirred whole blood frequently increases the rate and extent of the platelet aggregation that occurs; this is likely to be via immune complex formation between SK and natural anti-SK antibodies leading to increased release of pro-aggregatory factors.In this investigation we have examined the effects of several inhibitors and antagonists in an attempt to identify the proaggregatory factors that contribute to both SPA and SK-induced aggregation (SKA) and to evaluate different means of inhibiting both processes. The effects of the inhibitors/antagonists were determined in vitro after adding them to citrated whole blood obtained from healthy volunteers. Platelet aggregation was measured using a platelet counting technique.Inhibition of both SPA and SKA by apyrase and by FPL 66096 (a P2T receptor antagonist) demonstrated the involvement of ADP in both processes. Inhibition by chlorpromazine indicated that the most likely source of the ADP is red cells. The effects of sulotroban (a TXA2 antagonist) indicated involvement of TXA2 in SKA but not in SPA. The lack of effect of specific antagonists at S2, α2 and PAF receptors suggested lack of involvement of serotonin, catecholamines and plateletactivating factor in either SPA or SKA. Both SPA and SKA were potently inhibited by low concentrations of iloprost (a PGI2 analogue), but a high concentration of SIN-1 (a NO donor) was much less effective. SPA and SKA were prevented by EDTA and by RGDS indicating the importance of divalent cations and of the RGD sequence in adhesive proteins in mediating the platelet aggregation that occurred.We also determined the effects on SPA and SKA of adding MgCl2 to whole blood. In this case we used blood containing hirudin as anticoagulant. MgCl2 (1 mM) appeared to delay the onset of SPA and markedly inhibited SKA.


Gut ◽  
1997 ◽  
Vol 41 (6) ◽  
pp. 748-752 ◽  
Author(s):  
N Kalia ◽  
S Jacob ◽  
N J Brown ◽  
M W R Reed ◽  
D Morton ◽  
...  

Background—The exact mechanisms by whichHelicobacter pylori infection results in gastric mucosal injury are unclear.Aims—To assess in vivo whether H pylori extracts could initiate an inflammatory response in the rat gastric mucosal microcirculation.Methods—Extracts of H pylori, Escherichia coli, or distilled water were administered topically to the gastric mucosa of anaesthetised animals. Fluorescence in vivo microscopy assessed macromolecular leakage of labelled albumin from mucosal vessels, leucocyte adherence/rolling, and platelet activity for 90 minutes.Results—H pylori induced increases (p<0.001) in adherent platelet thrombi and circulating platelet emboli after five and 15 minutes respectively. Adherent platelet thrombi (mean of four per field of view) remained significantly increased throughout the experiment, but circulating emboli (maximum of five at 30 minutes) decreased with time. Leucocyte adherence did not occur although early transient rolling was observed. An 11% increase (p<0.02) in albumin leakage occurred after five minutes only. The induction of platelet aggregation was only observed following H pyloriadministration.Conclusion—This in vivo study demonstrated the ability of Hpylori extracts to promote platelet aggregation within gastric mucosal microvessels. Recruitment of leucocytes was not observed. The results suggest that the early events associated with H pylori infection are platelet aggregation with perhaps subsequent leucocyte recruitment by activated platelets.


Author(s):  
W. H. Zucker ◽  
R. G. Mason

Platelet adhesion initiates platelet aggregation and is an important component of the hemostatic process. Since the development of a new form of collagen as a topical hemostatic agent is of both basic and clinical interest, an ultrastructural and hematologic study of the interaction of platelets with the microcrystalline collagen preparation was undertaken.In this study, whole blood anticoagulated with EDTA was used in order to inhibit aggregation and permit study of platelet adhesion to collagen as an isolated event. The microcrystalline collagen was prepared from bovine dermal corium; milling was with sharp blades. The preparation consists of partial hydrochloric acid amine collagen salts and retains much of the fibrillar morphology of native collagen.


Sign in / Sign up

Export Citation Format

Share Document