Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cardiovasc Drugs Ther ; 37(5): 865-876, 2023 10.
Article in English | MEDLINE | ID: mdl-35595877

ABSTRACT

PURPOSE: The role of platelets during myocardial ischemia/reperfusion (I/R) is ambivalent. They contribute to injury but also to cardioprotection. Repeated blood flow restriction and reperfusion in a tissue/organ remote from the heart (remote ischemic conditioning, RIC) reduce myocardial I/R injury and attenuate platelet activation. Whether or not platelets mediate RIC's cardioprotective signal is currently unclear. METHODS AND RESULTS: Venous blood from healthy volunteers (without or with pretreatment of 500/1000 mg aspirin or 180 mg ticagrelor orally, 2-3 h before the study, n = 18 each) was collected before and after RIC (3 × 5 min blood pressure cuff inflation at 200 mmHg on the left upper arm/5 min deflation). Washed platelets were isolated. Platelet-poor plasma was used to prepare plasma-dialysates. Platelets (25 × 103/µL) or plasma-dialysates (1:10) prepared before and after RIC from untreated versus aspirin- or ticagrelor-pretreated volunteers, respectively, were infused into isolated buffer-perfused rat hearts. Hearts were subjected to global 30 min/120 min I/R. Infarct size was stained. Infarct size was less with infusion of platelets/plasma-dialysate after RIC (18 ± 7%/23 ± 9% of ventricular mass) than with platelets/plasma-dialysate before RIC (34 ± 7%/33 ± 8%). Aspirin pretreatment abrogated the transfer of RIC's cardioprotection by platelets (after/before RIC, 34 ± 7%/33 ± 7%) but only attenuated that by plasma-dialysate (after/before RIC, 26 ± 8%/32 ± 5%). Ticagrelor pretreatment induced an in vivo formation of cardioprotective factor(s) per se (platelets/plasma-dialysate before RIC, 26 ± 7%/26 ± 7%) but did not impact on RIC's cardioprotection by platelets/plasma-dialysate (20 ± 7%/21 ± 5%). CONCLUSION: Platelets serve as carriers for RIC's cardioprotective signal through an aspirin-sensitive and thus cyclooxygenase-dependent mechanism. The P2Y12 inhibitor ticagrelor per se induces a humoral cardioprotective signal.


Subject(s)
Aspirin , Ischemia , Rats , Animals , Humans , Ticagrelor/pharmacology , Aspirin/pharmacology , Infarction , Dialysis Solutions
3.
Circulation ; 130(19): 1700-11, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25239438

ABSTRACT

BACKGROUND: Diabetes mellitus predisposes to thrombotic and proliferative vascular remodeling, to which thrombin contributes via activation of protease-activated receptor (PAR) 1. However, the use of PAR-1 inhibitors to suppress remodeling may be limited by severe bleeding. We recently reported upregulation of an additional thrombin receptor, PAR-4, in human vascular smooth muscle cells exposed to high glucose and have now examined PAR-4 as a novel mediator linking hyperglycemia, hypercoagulation, and vascular remodeling in diabetes mellitus. METHODS AND RESULTS: PAR-4 expression was increased in carotid atherectomies and saphenous vein specimens from diabetic versus nondiabetic patients and in aorta and carotid arteries from streptozotocin-diabetic versus nondiabetic C57BL/6 mice. Vascular PAR-1 mRNA was not increased in diabetic mice. Ligated carotid arteries from diabetic mice developed more extensive neointimal hyperplasia and showed greater proliferation than arteries from nondiabetic mice. The augmented remodeling response was absent in diabetic mice deficient in PAR-4. At the cellular level, PAR-4 expression was controlled via the mRNA stabilizing actions of human antigen R, which accounted for the stimulatory actions of high glucose, angiotensin II, and H2O2 on PAR-4 expression, whereas cicaprost via protein kinase A activation counteracted this effect. CONCLUSIONS: PAR-4 appears to play a hitherto unsuspected role in diabetic vasculopathy. The development of PAR-4 inhibitors might serve to limit mainly proliferative processes in restenosis-prone diabetic patients, particularly those patients in whom severe bleeding attributed to selective PAR-1 blockade or complete thrombin inhibition must be avoided or those who do not require anticoagulation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/pathology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Atherectomy , Blood Glucose/metabolism , Carotid Artery Injuries/complications , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Female , Humans , Hyperglycemia/complications , Hyperglycemia/metabolism , Hyperglycemia/pathology , Ligation , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Saphenous Vein/cytology , Saphenous Vein/metabolism , Thrombin/metabolism , Thrombophilia/etiology , Thrombophilia/metabolism , Thrombophilia/pathology , Tunica Intima/metabolism , Tunica Intima/pathology
4.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 17-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26201059

ABSTRACT

Aspirin is an unique compound because it bears two active moieties within one and the same molecule: a reactive acetyl group and the salicylate metabolite. Salicylate has some effects similar to aspirin, however only at higher concentrations, usually in the millimolar range, which are not obtained at conventional antiplatelet aspirin doses of 100-300 mg/day. Pharmacological actions of aspirin in the cardiovascular system at these doses are largely if not entirely due to target structure acetylation. Several classes of lipid mediators become affected: Best known is the cyclooxygenase-1 (COX-1) in platelets with subsequent inhibition of thromboxane and, possibly, thrombin formation. By this action, aspirin also inhibits paracrine thromboxane functions on other lipid mediators, such as the platelet storage product sphingosine-1-phosphate (S1P), an inflammatory mediator. Acetylation of COX-2 allows for generation of 15-(R)HETE and subsequent formation of "aspirin-triggered lipoxin" (ATL) by interaction with white cell lipoxygenases. In the cardiovascular system, aspirin also acetylates eNOS with subsequent upregulation of NO formation and enhanced expression of the antioxidans heme-oxygenase-1. This action is possibly also COX-2/ATL mediated. Many more acetylation targets have been identified in live cells by quantitative acid-cleavable activity-based protein profiling and might result in discovery of even more aspirin targets in the near future.


Subject(s)
Aspirin/pharmacology , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Lipid Metabolism/drug effects , Aspirin/adverse effects , Aspirin/pharmacokinetics , Humans , Molecular Targeted Therapy
5.
Mediators Inflamm ; 2015: 831059, 2015.
Article in English | MEDLINE | ID: mdl-26604433

ABSTRACT

Sphingosine-1-phosphate (S1P) is a versatile lipid signaling molecule and key regulator in vascular inflammation. S1P is secreted by platelets, monocytes, and vascular endothelial and smooth muscle cells. It binds specifically to a family of G-protein-coupled receptors, S1P receptors 1 to 5, resulting in downstream signaling and numerous cellular effects. S1P modulates cell proliferation and migration, and mediates proinflammatory responses and apoptosis. In the vascular barrier, S1P regulates permeability and endothelial reactions and recruitment of monocytes and may modulate atherosclerosis. Only recently has S1P emerged as a critical mediator which directly links the coagulation factor system to vascular inflammation. The multifunctional proteases thrombin and FXa regulate local S1P availability and interact with S1P signaling at multiple levels in various vascular cell types. Differential expression patterns and intracellular signaling pathways of each receptor enable S1P to exert its widespread functions. Although a vast amount of information is available about the functions of S1P and its receptors in the regulation of physiological and pathophysiological conditions, S1P-mediated mechanisms in the vasculature remain to be elucidated. This review summarizes recent findings regarding the role of S1P and its receptors in vascular wall and blood cells, which link the coagulation system to inflammatory responses in the vasculature.


Subject(s)
Blood Coagulation/physiology , Inflammation/blood , Inflammation/immunology , Lysophospholipids/blood , Lysophospholipids/immunology , Receptors, Lysosphingolipid/blood , Receptors, Lysosphingolipid/immunology , Sphingosine/analogs & derivatives , Blood Coagulation/immunology , Blood Vessels/physiology , Endothelium, Vascular/physiology , Humans , Models, Cardiovascular , Models, Immunological , Platelet Activation , Receptors, Thrombin/metabolism , Signal Transduction , Sphingosine/blood , Sphingosine/immunology
6.
Biol Chem ; 394(6): 753-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23362192

ABSTRACT

Cyclooxygenase 1b (COX-1b) is a splice variant of COX-1, containing a retained intron 1 within the signal peptide sequence. COX-1b mRNA is found in many species, but the existence of a functionally active protein, which is possibly related to different species-dependent lengths of intron 1, is controversially discussed. The human intron 1 comprises 94 bp, and the resulting frameshift at the intron 1-exon 2 junction creates a premature stop codon. Nevertheless, full-length human COX-1b protein expression, including translated intron 1 and the signal peptide, has been reported and was explained by a frameshift repair. In this study, the fate of COX-1b mRNA in a human overexpression system is analyzed. Independent of the hypothetical frameshift repair mechanism, the splicing of the COX-1b intron 1, resulting in COX-1 mRNA and removal of the signal peptide during protein maturation, with subsequent generation of a COX-1 protein is demonstrated.


Subject(s)
Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Protein Biosynthesis , Amino Acid Sequence , Base Sequence , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , DNA, Complementary/genetics , Exons/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Introns/genetics , Liver/enzymology , Mass Spectrometry , Molecular Sequence Data , Prostaglandin-Endoperoxide Synthases/chemistry , Protein Sorting Signals , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Stomach/enzymology
7.
Med Monatsschr Pharm ; 36(11): 411-21, 2013 Nov.
Article in German | MEDLINE | ID: mdl-24640118

ABSTRACT

Observational but also some randomized trials suggest that regular long-term use of aspirin (acetylsalicylic acid) might reduce the risk of colorectal carcinomas by 15-40%. The efficacy appears to be increased with longer duration of treatment, i.e. beyond 5-10 years, but not with increasing doses. Aspirin at 75-100 mg daily appears to be sufficient in both primary as well as secondary prevention of recurrent tumors in sensitive persons, including prevention of distant metastases. The pharmacological mode of aspirin action is unclear as is the question whether only one or more sites of action exist. In any case, the mechanism(s) in charge should work at aspirin plasma levels of 10 microM or less which is the maximum concentration to be expected after antiplatelet doses. Inhibition of COX-1 and/or COX-2 is most likely involved. Follow-up reactions, such as inhibition of platelet-dependent thromboxane formation and action, release of storage products such as VEGF or sphingosine-1-phosphate and acetylation of COX-2 with subsequent generation of antioncogenic lipid mediators, such as lipoxins, are also possible. There is not much likelihood for "direct" actions of aspirin, shown in vitro at concentrations of 5 mM and more, which uncouple oxidative phosphorylation and paralyse the cell energy metabolism. benefit/risk profile, specifically regarding severe or fatal bleedings (GI-tract, cerebral). Accordingly, the actual German guideline "colorectal carcinoma" does not recommend aspirin use for prophylactic purposes. What is strongly needed are definitions of risk patients in terms of biomarkers or genetic profiling as well as data from long-term prospective randomized trials--both are underway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/therapeutic use , Colorectal Neoplasms/prevention & control , Platelet Aggregation Inhibitors/therapeutic use , Humans , Observational Studies as Topic , Primary Prevention , Randomized Controlled Trials as Topic , Risk Assessment , Secondary Prevention
8.
Thromb Haemost ; 123(2): 166-176, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36522182

ABSTRACT

Lipid-lowering agents and antiplatelet drugs are guideline-recommended standard treatment for secondary prevention of acute thrombotic events in patients with increased cardiovascular risk. Aspirin is the most frequently used antiplatelet drug, either alone or in combination with other antiplatelet agents (P2Y12 inhibitors), while statins are first-line treatment of hypercholesterolemia. The well-established mode of action of aspirin is inhibition of platelet-dependent thromboxane formation. In addition, aspirin also improves endothelial oxygen defense via enhanced NO formation and inhibits thrombin formation. Low-dose aspirin exerts in addition anti-inflammatory effects, mainly via inhibition of platelet-initiated activation of white cells.Statins inhibit platelet function via reduction of circulating low-density lipoprotein-cholesterol (LDL-C) levels and a more direct inhibition of platelet function. This comprises inhibition of thromboxane formation via inhibition of platelet phospholipase A2 and inhibition of (ox)LDL-C-mediated increases in platelet reactivity via the (ox)LDL-C receptor (CD36). Furthermore, statins upregulate endothelial NO-synthase and improve endothelial oxygen defense by inhibition of NADPH-oxidase. PCSK9 antibodies target a serine protease (PCSK9), which promotes the degradation of the LDL-C receptor impacting on LDL-C plasma levels and (ox)LDL-C-receptor-mediated signaling in platelets similar to but more potent than statins.These functionally synergistic actions are the basis for numerous interactions between antiplatelet and these lipid-lowering drugs, which may, in summary, reduce the incidence of atherothrombotic vascular events.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Aspirin , Cholesterol, LDL , Drug Interactions , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypolipidemic Agents/therapeutic use , PCSK9 Inhibitors , Platelet Aggregation Inhibitors/therapeutic use , Proprotein Convertase 9/metabolism , Thromboxanes
9.
Hepatology ; 54(1): 204-15, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21452284

ABSTRACT

UNLABELLED: Astrocytes play an important role in the pathogenesis of hepatic encephalopathy (HE) and ammonia toxicity, whereas little is known about microglia and neuroinflammation under these conditions. We therefore studied the effects of ammonia on rat microglia in vitro and in vivo and analyzed markers of neuroinflammation in post mortem brain tissue from patients with cirrhosis with and without HE and non-cirrhotic controls. In cultured rat microglia, ammonia stimulated cell migration and induced oxidative stress and an up-regulation of the microglial activation marker ionized calcium-binding adaptor molecule-1 (Iba-1). Up-regulation of Iba-1 was also found in the cerebral cortex from acutely ammonia-intoxicated rats and in the cerebral cortex from patients with cirrhosis who have HE, but not from patients with cirrhosis who do not have HE. However, ammonia had no effect on microglial glutamate release, prostaglandin synthesis, and messenger RNA (mRNA) levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and the proinflammatory cytokines interleukin (IL)-1α/ß, tumor necrosis factor α, or IL-6, whereas in cultured astrocytes ammonia induced the release of glutamate, prostaglandins, and increased IL-1ß mRNA. mRNA and protein expression of iNOS and COX-2 or mRNA expression of proinflammatory cytokines and chemokine monocyte chemoattractive protein-1 in cerebral cortex from patients with liver cirrhosis and HE were not different from those found in patients with cirrhosis who did not have HE or control patients without cirrhosis. CONCLUSION: These data suggest that microglia become activated in experimental hyperammonemia and HE in humans and may contribute to the generation of oxidative stress. However, HE in patients with liver cirrhosis is not associated with an up-regulation of inflammatory cytokines in cerebral cortex, despite microglia activation.


Subject(s)
Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Microglia/metabolism , Microglia/pathology , Ammonia/adverse effects , Ammonia/pharmacology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Autopsy , Calcium-Binding Proteins/metabolism , Cell Movement/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Hepatic Encephalopathy/chemically induced , Humans , Male , Microfilament Proteins/metabolism , Oxidative Stress/drug effects , Phagocytosis/drug effects , Rats , Rats, Wistar
10.
Arterioscler Thromb Vasc Biol ; 31(3): 624-33, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21164077

ABSTRACT

OBJECTIVE: Diabetes is associated with vascular remodeling and increased thrombin generation. Thrombin promotes vascular smooth muscle cell (SMC) mitogenesis and migration via protease-activated receptors (PAR)-1, PAR-3, and PAR-4. We investigated the effect of high glucose on expression and function of vascular thrombin receptors. METHODS AND RESULTS: In human vascular SMCs, high glucose (25 versus 5.5 mmol/L) induced a rapid and sustained increase in PAR-4 mRNA, protein, and cell surface expression. PAR-1 and PAR-3 expression were not changed. High glucose pretreatment (48 hours) enhanced thrombin or PAR-4-activating peptide but not PAR-1-activating peptide evoked intracellular calcium mobilization, migration, and tumor necrosis factor α gene expression. This enhancement of thrombin-stimulated migration and gene expression by high glucose was abolished by endogenous PAR-4 knockdown. PAR-4 regulation was prevented by inhibition of protein kinase (PK)C-ß and -δ isoforms or nuclear factor (NF)κB. Nuclear translocation of NFκB in high glucose-stimulated SMCs led to PKC-dependent NFκB binding to the PAR-4 promoter in a chromatin immunoprecipitation assay. Furthermore, in situ hybridization and immunohistochemistry confirmed high abundance of PAR-4 in human diabetic vessels as compared with nondiabetic vessels. CONCLUSIONS: High glucose enhances SMC responsiveness to thrombin through transcriptional upregulation of PAR-4, mediated via PKC-ß, -δ, and NFκB. This may play an important role in the vascular complications of diabetes.


Subject(s)
Calcium Signaling , Diabetic Angiopathies/metabolism , Glucose/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Binding Sites , Calcium/metabolism , Cell Movement , Cells, Cultured , Chromatin Immunoprecipitation , Diabetic Angiopathies/genetics , Humans , Immunohistochemistry , In Situ Hybridization , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Messenger/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/genetics , Time Factors , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
11.
Mol Pharmacol ; 80(2): 337-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21596928

ABSTRACT

Thrombin promotes vascular smooth muscle cell (SMC) proliferation and inflammation via protease-activated receptor (PAR)-1. A further thrombin receptor, PAR-3, acts as a PAR-1 cofactor in some cell-types. Unlike PAR-1, PAR-3 is dynamically regulated at the mRNA level in thrombin-stimulated SMC. This study investigated the mechanisms controlling PAR-3 expression. In human vascular SMC, PAR-3 siRNA attenuated thrombin-stimulated interleukin-6 expression and extracellular signal-regulated kinases 1/2 phosphorylation, indicating PAR-3 contributes to net thrombin responses in these cells. Thrombin slowed the decay of PAR-3 but not PAR-1 mRNA in the presence of actinomycin D and induced cytosolic shuttling and PAR-3 mRNA binding of the mRNA-stabilizing protein human antigen R (HuR). HuR siRNA prevented thrombin-induced PAR-3 expression. By contrast, forskolin inhibited HuR shuttling and destabilized PAR-3 mRNA, thus reducing PAR-3 mRNA and protein expression. Other cAMP-elevating agents, including the prostacyclin-mimetic iloprost, also down-regulated PAR-3, accompanied by decreased HuR/PAR-3 mRNA binding. Iloprost-induced suppression of PAR-3 was reversed with a myristoylated inhibitor of protein kinase A and mimicked by phorbol ester, an inducer of cyclooxygenase-2. In separate studies, iloprost attenuated PAR-3 promoter activity and prevented binding of nuclear factor of activated T cells (NFAT2) to the human PAR-3 promoter in a chromatin immunoprecipitation assay. Accordingly, PAR-3 expression was suppressed by the NFAT inhibitor cyclosporine A or NFAT2 siRNA. Thus human PAR-3, unlike PAR-1, is regulated post-transcriptionally via the mRNA-stabilizing factor HuR, whereas transcriptional control involves NFAT2. Through modulation of PAR-3 expression, prostacyclin and NFAT inhibitors may limit proliferative and inflammatory responses to thrombin after vessel injury.


Subject(s)
Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors/physiology , RNA Stability/genetics , RNA, Messenger/genetics , Receptors, Thrombin/physiology , Antigens, Surface/genetics , Antigens, Surface/physiology , Cyclic AMP/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Gene Knockdown Techniques/methods , Humans , NFATC Transcription Factors/genetics , Oligopeptides/genetics , Oligopeptides/physiology , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Receptors, Thrombin/genetics
12.
Arterioscler Thromb Vasc Biol ; 30(12): 2434-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21071695

ABSTRACT

OBJECTIVE: The platelet P2Y12 ADP receptor is a well-known target of thienopyridine-type antiplatelet drugs. This study is the first to describe increased transcriptional expression of a functionally active P2Y12 in response to thrombin in human vascular smooth muscle cells (SMC). METHODS AND RESULTS: On exposure to thrombin, P2Y12 mRNA was transiently increased, whereas total protein and cell surface expression of P2Y12 were markedly increased within 6 hours and remained elevated over 24 hours. This effect was mediated by activation of nuclear factor κB. Preincubation with thrombin significantly enhanced the efficacy of the P2Y receptor agonist 2-methylthio-ADP to induce interleukin 6 expression and SMC mitogenesis. Effects induced by 2-methylthio-ADP were prevented by RNA interference-mediated knockdown of P2Y12 and a selective P2Y12-antagonist R-138727, the active metabolite of prasugrel. In addition, positive P2Y12 immunostaining was shown in SMC of human carotid artery plaques and was found to colocalize with tissue factor, the rate-limiting factor of thrombin formation in vivo. CONCLUSIONS: These data suggest that the P2Y12 receptor not only is central to ADP-induced platelet activation but also may mediate platelet-independent responses, specifically under conditions of enhanced thrombin formation, such as local vessel injury and atherosclerotic plaque rupture.


Subject(s)
Adenosine Diphosphate/metabolism , Carotid Artery Diseases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Purinergic P2Y12/metabolism , Thrombin/metabolism , Transcriptional Activation , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Carotid Artery Diseases/pathology , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Humans , Interleukin-6/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Piperazines/pharmacology , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , RNA Interference , RNA, Messenger/metabolism , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/genetics , Thionucleotides/pharmacology , Time Factors , Transcriptional Activation/drug effects , Up-Regulation
13.
J Exp Pharmacol ; 13: 957-970, 2021.
Article in English | MEDLINE | ID: mdl-34908882

ABSTRACT

INTRODUCTION: Pharmacologic therapy options for COVID-19 should include antiviral, anti-inflammatory, and anticoagulant agents. With the limited effectiveness, currently available virus-directed therapies may have a substantial impact on global health due to continued reports of mutant variants affecting repeated waves of COVID-19 around the world. METHODS: We searched articles pertaining to aspirin, COVID-19, acute lung injury and pharmacology in PubMed and provide a comprehensive appraisal of potential use of aspirin in the management of patients with COVID-19. The scope of this article is to provide an overview of the rationale and currently available clinical evidence that supports aspirin as an effective therapeutic option in COVID-19. RESULTS: Experimental and clinical evidence are available for the potential use of aspirin in patients with COVID-19. DISCUSSION: Aspirin targets the intracellular signaling pathway that is essential for viral replication, and resultant inflammatory responses, hypercoagulability, and platelet activation. With these multiple benefits, aspirin can be a credible adjunctive therapeutic option for the treatment of COVID-19. In addition, inhaled formulation with its rapid effects may enhance direct delivery to the lung, which is the key organ damaged in COVID-19 during the critical initial course of the disease, whereas the 150-325 mg/day can be used for long-term treatment to prevent thrombotic event occurrences. Being economical and widely available, aspirin can be exploited globally, particularly in underserved communities and remote areas of the world to combat the ongoing COVID-19 pandemic.

14.
J Am Heart Assoc ; 10(22): e022299, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34726072

ABSTRACT

Background Pain is a major issue in our aging society. Dipyrone (metamizole) is one of the most frequently used analgesics. Additionally, it has been shown to impair pharmacodynamic response to aspirin as measured by platelet function tests. However, it is not known how this laboratory effect translates to clinical outcome. Methods and Results We conducted a nationwide analysis of a health insurance database in Germany comprising 9.2 million patients. All patients with a cardiovascular event in 2014 and subsequent secondary prevention with aspirin were followed up for 36 months. Inverse probability of treatment weighting analysis was conducted to investigate the rate of mortality, myocardial infarction, and stroke/transient ischemic attack between patients on aspirin-dipyrone co-medication compared with aspirin-alone medication. Permanent aspirin-alone medication was given to 26,200 patients, and 5946 patients received aspirin-dipyrone co-medication. In the inverse probability of treatment weighted sample, excess mortality in aspirin-dipyrone co-medicated patients was observed (15.6% in aspirin-only group versus 24.4% in the co-medicated group, hazard ratio [HR], 1.66 [95% CI, 1.56-1.76], P<0.0001). Myocardial infarction and stroke/transient ischemic attack were increased as well (myocardial infarction: 1370 [5.2%] versus 355 [5.9%] in aspirin-only and co-medicated groups, respectively; HR, 1.18 [95% CI, 1.05-1.32]; P=0.0066, relative risk [RR], 1.14; number needed to harm, 140. Stroke/transient ischemic attack, 1901 [7.3%] versus 506 [8.5%] in aspirin-only and co-medicated groups, respectively; HR, 1.22 [95% CI, 1.11-1.35]; P<0.0001, RR, 1.17, number needed to harm, 82). Conclusions In this observational, nationwide analysis, aspirin and dipyrone co-medication was associated with excess mortality. This was in part driven by ischemic events (myocardial infarction and stroke), which occurred more frequently in co-medicated patients as well. Hence, dipyrone should be used with caution in aspirin-treated patients for secondary prevention.


Subject(s)
Aspirin/adverse effects , Cardiovascular Diseases , Dipyrone/adverse effects , Cardiotoxins , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Humans , Ischemic Attack, Transient/diagnosis , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/epidemiology , Myocardial Infarction/drug therapy , Myocardial Infarction/epidemiology , Platelet Aggregation Inhibitors/adverse effects , Stroke/diagnosis , Stroke/epidemiology , Stroke/prevention & control
15.
Glia ; 58(6): 691-705, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20014275

ABSTRACT

Ammonia toxicity to the brain involves NMDA receptor overactivation and glutamate excitotoxicity. The mechanisms underlying glutamate release from astrocytes in response to ammonia were addressed in this study. In cultured rat astrocytes, glutamate immunoreactivity (IR) was punctate and partly colocalized with transfected VAMP2-YFP. NH(4)Cl (5 mmol/L) and hypoosmotic exposure (205 mosmol/L) induced a rapid colchicine-sensitive loss of cellular glutamate and glutamate appearance in the extracellular space. The NH(4)Cl-induced glutamate loss from astrocytes was strongly blunted after transfection of the cells with VAMP2 siRNA. Ammonia-induced exocytosis of VAMP2-YFP expressing vesicles was shown by total internal reflection fluorescence microscopy (TIRF-M). Glutamate exocytosis in response to ammonia was sensitive to chelation of Ca(2+), cyclooxygenase inhibition by indomethacin and colchicine. Ammonia triggered the rapid formation of prostanoids, which were identified as upstream events in ammonia-induced glutamate exocytosis. Also, addition of prostaglandin E(2) or of tumor necrosis factor (TNF)-alpha triggered glutamate exocytosis. Inhibition of ammonia-induced glutamate exocytosis after transfection of VAMP2 siRNA inhibited ammonia-induced RNA oxidation. It is concluded that ammonia triggers a prostanoid- and Ca(2+)-dependent glutamate exocytosis, which is essential for induction of ammonia-induced RNA oxidation.


Subject(s)
Ammonia/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Exocytosis/drug effects , Glutamic Acid/metabolism , Animals , Animals, Newborn , Bacterial Proteins/genetics , Brain/cytology , Calcium/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Fluorometry/methods , Glial Fibrillary Acidic Protein/metabolism , Indomethacin/pharmacology , Luminescent Proteins/genetics , Prostaglandins/metabolism , Protein Transport/drug effects , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , RNA/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Transfection/methods , Vesicle-Associated Membrane Protein 2/metabolism
16.
Basic Res Cardiol ; 105(4): 479-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20177692

ABSTRACT

Clinical studies revealed unfavorable effects of hormone replacement therapy in postmenopausal women despite strong evidence for vasoprotective effects of estrogen in animal models. Therefore, an attempt was made to address adverse effects of estradiol on atherosclerosis, endothelial function, and thrombosis in a murine model of atherosclerosis. ApoE(-/-) mice were bilaterally ovariectomized (OVX) and substituted with placebo or 17-beta-Estradiol (E(2), 1.1 and 6.6 microg/day) on Western diet for 90 days. Low-dose E(2) (1.1 microg/day) treatment significantly increased atherosclerotic plaque score, whereas high-dose E(2) (6.6 microg/day) reduced aortic plaque burden. The proatherosclerotic effects of low-dose E(2) were associated with decreased total collagen in aortic root lesions and impaired acetylcholine (ACh)-induced vasorelaxation of aortic rings. On the contrary, OVX compared with control reduced atherosclerosis, increased fibrillar collagen and improved endothelial function. The thrombotic response as measured in a photothrombosis model was not significantly altered by E(2) or OVX. Taken together, differential effects on atherosclerosis of the clinical relevant low-dose E(2) compared with high-dose E(2) were demonstrated. Importantly, the presented experimental conditions provide a model to study the untoward vascular effects of E(2) in the context of accelerated and advanced atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Estradiol/metabolism , Animals , Aorta/physiology , Atherosclerosis/etiology , Collagen/metabolism , Dietary Fats/adverse effects , Endothelium, Vascular/physiology , Female , Mice , Mice, Knockout , Ovariectomy , Vasodilation
17.
Circ Res ; 100(2): 255-62, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17170365

ABSTRACT

The endothelial cell protein C receptor (EPCR) is expressed on endothelial cells and regulates the protein C anticoagulant pathway via the thrombin-thrombomodulin complex. Independent of its anticoagulant activity, activated protein C (APC) can directly signal to endothelial cells and upregulate antiapoptotic and antiinflammatory genes. Here we show that vascular smooth muscle cells (SMCs) also express EPCR. EPCR protein on SMCs was detected by flow cytometry and Western blotting. EPCR mRNA was identified by quantitative RT-PCR. To examine the functionality of EPCR, intracellular signaling in APC-stimulated SMCs was analyzed by determination of intracellular free calcium transients using confocal laser scanning microscopy. Phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK-1/2) was detected by immunoblotting. APC-induced ERK-1/2 phosphorylation was inhibited by an anti-EPCR antibody and by a cleavage site blocking anti-PAR-1 antibody, indicating that binding of APC to EPCR and cleavage of protease-activated receptor-1 (PAR-1) were involved. APC elicited an increase in [(3)H]-thymidine incorporation. The mitogenic effect of APC was significantly enhanced in the presence of thrombin. EPCR expression was also detected in SMCs in the fibrous cap of human carotid artery plaques. The present data demonstrate functionally active EPCR in SMCs and suggest that EPCR-bound APC might modulate PAR-1-mediated responses of SMCs to vascular injury.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein C/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Antigens, CD/physiology , Carotid Stenosis/enzymology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cells, Cultured , Coculture Techniques , Endothelial Protein C Receptor , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Humans , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Protein Binding/physiology , Receptors, Cell Surface/physiology
18.
Arterioscler Thromb Vasc Biol ; 28(3): 534-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18162607

ABSTRACT

OBJECTIVE: Stimulation of protease-activated receptor-1 (PAR-1) by thrombin causes vascular smooth muscle cell (SMC) mitogenesis and has been implicated in the vascular response to injury. Vascular injury is also associated with enhanced formation of PGE2 and PGI2 (prostacyclin). This study investigates whether PGI2 and PGE2 modify the expression of PAR-1 and the cellular response to thrombin in human SMC. METHODS AND RESULTS: The PGI2-mimetic iloprost (1 to 100 nmol/L) attenuated mRNA, total protein, and cell surface expression of PAR-1. This was associated with inhibition of thrombin-induced mitogenesis and migration. Comparable inhibition of PAR-1 expression was observed with the selective IP-receptor agonist cicaprost, the adenylyl cyclase activator forskolin, the phosphodiesterase inhibitor isobutylmethylxanthine and the PKA activator dibutyryl-cAMP. Similar effects of PGE2 required micromolar concentrations. The specific PKA-inhibitor Myr-PKI prevented PAR-1 downregulation by iloprost. The potential role of Rho family GTPases in PAR-1 regulation was also investigated. Iloprost decreased Rac1 mRNA and the Rac1 inhibitor NSC23766 mimicked the inhibitory effects of iloprost on PAR-1 protein--but not mRNA. The Rho kinase inhibitor Y27632 did not influence PAR-1 expression. CONCLUSIONS: IP-receptor agonists may limit the mitogenic actions of thrombin in human SMC by downregulating PAR-1 via modulation of cAMP-/PKA- and Rac1-dependent signaling pathways.


Subject(s)
Epoprostenol/physiology , Iloprost/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, PAR-1/metabolism , Transcription, Genetic/physiology , Analysis of Variance , Blotting, Western , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Dinoprostone/pharmacology , Dinoprostone/physiology , Down-Regulation , Epoprostenol/pharmacology , Flow Cytometry , Gene Expression , Humans , Iloprost/pharmacology , Immunohistochemistry , Muscle, Smooth, Vascular/cytology , Probability , RNA, Messenger/analysis , Receptor, PAR-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein/cytology , Signal Transduction/genetics , Signal Transduction/physiology
19.
Arterioscler Thromb Vasc Biol ; 28(7): 1375-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18451333

ABSTRACT

OBJECTIVE: Apoptotic monocytes release membrane microparticles which may play a major role in thrombogenicity through a P-selectin glycoprotein ligand (PGSL-1)-mediated mechanism. We have studied systematically the regulation of PSGL-1 expression and function in apoptotic monocytic cells. METHODS AND RESULTS: PSGL-1 expression (flow cytometry, immunofluorescence microscopy, immunoblot) was virtually abolished in apoptotic monocytes by proteolytic shedding. This was accompanied by a complete loss of PSGL-1-mediated platelet-leukocyte (flow cytometry) and leukocyte-endothelial cell (parallel plate flow chamber) interactions. Systematic screening of protease inhibitors combined with knock-out and siRNA experiments characterized the PSGL-1-cleaving enzyme as an N-ethylmaleimide-inhibitable metalloproteinase of the ADAM family. CONCLUSIONS: Downmodulation of PGSL-1 in apoptotic monocytes may prevent ectopic cell clearance in the peripheral vasculature to reduce local inflammatory and proliferative responses. Depletion of PSGL-1 expression on apoptotic microparticles may also act as a molecular switch to modulate their thrombogenic activity.


Subject(s)
ADAM Proteins/metabolism , Apoptosis , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Transport Vesicles/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , Animals , Apoptosis/drug effects , Cell Adhesion , Down-Regulation , Endothelial Cells/metabolism , Fibroblasts/metabolism , Humans , Leukocytes/metabolism , Membrane Glycoproteins/genetics , Mice , Monocytes/drug effects , Monocytes/enzymology , Monocytes/pathology , Protease Inhibitors/pharmacology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Time Factors , Transfection , Transport Vesicles/drug effects , U937 Cells
20.
J Leukoc Biol ; 83(6): 1379-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18305177

ABSTRACT

Monocyte apoptosis is an important determinant of atherothrombosis. Two major mechanisms for apoptosis-associated thrombogenicity have been described: exposure of negatively charged membrane phospholipids and up-regulation of tissue factor (TF). However, the relative importance of these mechanisms is unclear. Thus, procoagulant functions (thrombin generation) of apoptotic (staurosporine, 2 muM, 24 h) U937 cells versus cell-derived microparticles (MPs) were studied. In apoptotic U937 cells, a significant increase in TF mRNA (real-time PCR), surface expression of TF (flow cytometry), and total cellular amount of TF (Western blotting) was observed. Control cells only minimally triggered thrombin generation (endogenous thrombin potential), and apoptotic cells were highly procoagulant. However, addition of negatively charged membranes completely restored the thrombin generation capacity of control U937 cells to the levels of apoptotic cells. MPs (defined as CD45(+) particles of subcellular size), derived from apoptotic U937 cells, were highly procoagulant but did not exhibit an increased TF expression or annexin V binding. Taken together, our data support the concept that the membrane environment, independent of TF expression, determines the extent of thrombin formation triggered by apoptosis of monocytic cells. Externalization of negatively charged phospholipids represents the most important mechanisms for whole cells. Additional yet unknown mechanisms appear to be involved in the procoagulant actions of MPs derived from apoptotic monocytes.


Subject(s)
Apoptosis , Monocytes/physiology , Thrombin/biosynthesis , Thromboplastin/physiology , Blood Coagulation Factors/physiology , Cell Membrane/metabolism , Humans , Staurosporine/pharmacology , Thromboplastin/genetics , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL