Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.288
Filter
1.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576080

ABSTRACT

Aside from the established immune-mediated etiology of multiple sclerosis (MS), compelling evidence implicates platelets as important players in disease pathogenesis. Specifically, numerous studies have highlighted that activated platelets promote the central nervous system (CNS)-directed adaptive immune response early in the disease course. Platelets, therefore, present a novel opportunity for modulating the neuroinflammatory process that characterizes MS. We hypothesized that the well-known antiplatelet agent acetylsalicylic acid (ASA) could inhibit neuroinflammation by affecting platelets if applied at low-dose and investigated its effect during experimental autoimmune encephalomyelitis (EAE) as a model to study MS. We found that oral administration of low-dose ASA alleviates symptoms of EAE accompanied by reduced inflammatory infiltrates and less extensive demyelination. Remarkably, the percentage of CNS-infiltrated CD4+ T cells, the major drivers of neuroinflammation, was decreased to 40.98 ± 3.28% in ASA-treated mice compared to 56.11 ± 1.46% in control animals at the disease maximum as revealed by flow cytometry. More interestingly, plasma levels of thromboxane A2 were decreased, while concentrations of platelet factor 4 and glycoprotein VI were not affected by low-dose ASA treatment. Overall, we demonstrate that low-dose ASA could ameliorate the platelet-dependent neuroinflammatory response in vivo, thus indicating a potential treatment approach for MS.


Subject(s)
Aspirin/pharmacology , Blood Platelets/pathology , Brain/pathology , Inflammation/pathology , Multiple Sclerosis/blood , Multiple Sclerosis/pathology , Platelet Aggregation Inhibitors/pharmacology , Animals , Aspirin/administration & dosage , Aspirin/therapeutic use , Blood Platelets/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/immunology , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Thromboxane A2/biosynthesis
3.
Sci Rep ; 10(1): 21420, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293599

ABSTRACT

Neointima hyperplasia is a crucial component of restenosis after coronary angioplasty. We have hypothesized that enhanced generation of platelet-derived thromboxane (TX)A2 in response to vascular damage plays a critical role in neointimal hyperplasia and that antiplatelet agents may mitigate it. In cocultures of human platelets and coronary artery smooth muscle cells (CASMC), we found that platelets induced morphologic changes and enhanced the migration of CASMC. The exposure of platelets to Aspirin [an inhibitor of cyclooxygenase (COX)-1] reduced the generation of TXA2 and prevented the morphological and functional changes induced by platelets in CASMC. Platelet-derived TXA2 induced COX-2 and enhanced prostaglandin (PG)E2 biosynthesis in CASMC, a known mechanism promoting neointimal hyperplasia. COX-2 induction was prevented by different antiplatelet agents, i.e., Aspirin, the TP antagonist SQ29,548, or Revacept (a dimeric soluble GPVI-Fc fusion protein). The administration of the novel antiplatelet agent Revacept to C57BL/6 mice, beginning three days before femoral artery denudation, and continuing up to seven days after injury, prevented the increase of the systemic biosynthesis di TXA2 and reduced femoral artery intima-to-media area and the levels of markers of cell proliferation and macrophage infiltration. Revacept might serve as a therapeutic agent for percutaneous coronary angioplasty and stent implantation.


Subject(s)
Blood Platelets/cytology , Coronary Vessels/cytology , Glycoproteins/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Neointima/prevention & control , Platelet Aggregation Inhibitors/pharmacology , Thromboxane A2/biosynthesis , Urine/chemistry , Adult , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Cyclooxygenase 2/metabolism , Humans , Hyperplasia , Male , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Neointima/metabolism , Neointima/pathology , Young Adult
4.
Biol Pharm Bull ; 43(8): 1283-1287, 2020.
Article in English | MEDLINE | ID: mdl-32741952

ABSTRACT

Although vascular dysfunction is a key event in the development of diabetic complications, and abnormal toll-like receptor 4 (TLR4) may contribute to the pathophysiology of vascular diseases, the direct relationships between TLR4 and vascular function in diabetic arteries are still poorly understood. Thus, to investigate whether pharmacological blockade of TLR4 affects vascular function in the superior mesenteric artery (SMA) of streptozotocin (STZ)-induced diabetic rats, the SMA was isolated from male Wistar rat injected once with STZ (65 mg/kg, 27-34 weeks) which was treated with TAK-242 (10-6 M), a TLR4 inhibitor, for approximately 1 d using organ culture techniques. After incubation, functional and biochemical studies were performed. In the functional study, treatment with TAK-242 increased acetylcholine (ACh)-induced relaxation of the diabetic SMA in the intact condition. Sodium nitroprusside (SNP)-induced relaxation was also increased in the TAK-242-treated group compared with the vehicle-treated group. Under cyclooxygenase (COX) blockade by indomethacin (10-5 M), ACh-induced relaxation was similar in the vehicle- and TAK-242-treated groups. In addition, ACh-induced relaxation in the combined presence of the nitric oxide (NO) synthase inhibitor, NG-nitro-L-arginine (L-NNA) (10-4 M), and indomethacin (10-5 M) was similar in the vehicle- and TAK-242-treated groups. The productions of thromboxane (TX) B2 in cultured medium in the presence of ACh (10-5 M) were lower in the TAK-242-treated group than in the vehicle-treated group. These data suggested that TAK-242 could augment endothelium-dependent relaxation by partly suppressing vasoconstrictor TXA2 or increasing NO signaling. TLR4 inhibition may be a novel therapeutic strategy to assist in the management of diabetes-associated vascular complications.


Subject(s)
Acetylcholine/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/drug therapy , Mesenteric Artery, Superior/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Vasodilation/drug effects , Animals , Endothelium-Dependent Relaxing Factors/physiology , Male , Mesenteric Artery, Superior/physiopathology , Rats , Rats, Wistar , Streptozocin , Sulfonamides/therapeutic use , Thromboxane A2/biosynthesis
5.
Biomed Pharmacother ; 130: 110535, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32707441

ABSTRACT

Pathophysiological reaction of platelets in the blood vessel is an indispensable part of thrombosis and cardiovascular disease, which is the most common cause of death in the world. In this study, we performed in vitro assays to evaluate antiplatelet activity of arctigenin in human platelets and attempted to identify the mechanism responsible for thromboxane A2 (TXA2) generation, integrin αIIbß3 activation and cAMP pathway. Arctigenin exhibited obvious inhibitory effects on collagen-, thrombin-, and ADP-induced human platelet aggregation, granule secretion, TXA2 generation, integrin αIIbß3 activation, and clot retraction. Additionally, we found that arctigenin attenuated PI3K/Akt/mTOR/GSK-3ß and MAPK signaling pathways, and increased cAMP level. Accordingly, the findings support that arctigenin attenuates thrombotic events through the inhibition of platelet activation and platelet plug formation. Therefore, we suggest that arctigenin may have therapeutic potential as an antiplatelet and antithrombotic agent.


Subject(s)
Clot Retraction/drug effects , Cyclic AMP , Fibrinolytic Agents/pharmacology , Furans/pharmacology , Lignans/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/drug effects , Thromboxane A2/biosynthesis , Blood Platelets/drug effects , Glycogen Synthase Kinase 3 beta/drug effects , Humans , In Vitro Techniques , Mitogen-Activated Protein Kinases , Oncogene Protein v-akt/drug effects , Phosphatidylinositol 3-Kinases/drug effects , TOR Serine-Threonine Kinases/drug effects
6.
Med Chem ; 16(1): 63-68, 2020.
Article in English | MEDLINE | ID: mdl-30734682

ABSTRACT

BACKGROUND: A convenient approach to modulation of the inflammation has an influence on the production of inflammatory mediators - icosanoids, generated in arachidonic acid (AA) metabolism. The common therapeutic activity of non-steroidal anti-inflammatory drugs (NSAID), such as aspirin, includes inhibition of two crucial enzymes of AA metabolism - cyclooxygenase- 1 and -2 (COX-1/2), with certain risk for gastrointestinal and renal intolerance. Ever since the enrolment of COX-2, particularly overabundance of its main products prostaglandin E2 (PGE2) and thromboxane A2 (TXA2) in numerous pathological processes was recognized, it became a significant therapeutic target. OBJECTIVE: The aim of this study was to examine the effects of synthesized organo-fluorine compounds on PGE2 and TXA2 production in the inflammation process. METHODS: Trifluoromethyl compounds were synthesized from N-benzyl trifluoromethyl aldimine, commercially available 2-methyl or 2-phenyl α-bromo esters (ß-lactams trans-1 and trans-2 and trifluoromethyl ß-amino ester, respectively) and methyl 2-isocyanoacetate (2-imidazoline trans-4). The reactions proceeded with high geometric selectivity, furnishing the desired products in good yields. The influence of newly synthesized compounds on PGE2 and TXA2 production in human leukemic U937 macrophages on both enzyme activity and gene expression levels was observed. RESULTS: Among the tested trifluoromethyl compounds, methyl trans-1-benzyl-5-(trifluoromethyl)- 4,5-dihydro-1H-imidazole-4-carboxylate (trans-4) can be distinguished as the most powerful antiinflammatory agent, probably due to its trifluoromethyl-imidazoline moiety. CONCLUSION: Some further structural modifications in tested compounds and particularly in the synthesis of different trifluoromethyl imidazolines could contribute to the development of new COX-2 inhibitors and potent anti-inflammatory agents.


Subject(s)
Dinoprostone/biosynthesis , Hydrocarbons, Fluorinated/pharmacology , Macrophages/drug effects , Thromboxane A2/biosynthesis , Dose-Response Relationship, Drug , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/metabolism , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured , U937 Cells
7.
J Thromb Haemost ; 17(6): 885-895, 2019 06.
Article in English | MEDLINE | ID: mdl-30933424

ABSTRACT

BACKGROUND: The prevalence and degree of obesity is rising worldwide, increases cardiovascular risk, modifies body composition and organ function, and potentially affects the pharmacokinetics and/or pharmacodynamics of drugs. OBJECTIVES: To investigate the pharmacodynamics of once-daily low-dose aspirin in healthy obese subjects, and to assess whether body weight (BW) and body mass index (BMI) affect the pharmacology of aspirin. PATIENTS/METHODS: Otherwise healthy, obese (BMI > 30 kg/m2 ) subjects were studied before and after 3-4 weeks of 100-mg once-daily aspirin intake. Aspirin pharmacodynamics were assessed according to serum thromboxane (TX) B2 levels measured at 4 hours, 24 hours (i.e., posologic interval) and 48 hours after the last witnessed intake; age-matched and sex-matched non-obese controls were included. A previously calibrated pharmacokinetic/pharmacodynamic in silico model of aspirin was used to fit serum TXB2 data from obese subjects. At baseline, the major urinary TXA2 and prostacyclin metabolites, urinary isoprostane and plasma inflammatory biomarkers were measured. RESULTS: In 16 obese subjects (aged 47 ± 11 years; BMI of 39.4 ± 5.1 kg/m2 ), residual serum TXB2 values between 4 and 48 hours after aspirin intake were increased 3- to 5-fold as compared with controls. At 24 hours, the residual serum TXB2 level was log-linearly associated with body size over a wide range of BMI and BW values, without any apparent threshold. The in silico model predicted that reduced aspirin bioavailability would be inversely related to body size and rescued by 200 mg of aspirin once daily or 85 mg twice daily. Baseline urinary TXA2 metabolite, isoprostane and plasma C-reactive protein levels were significantly increased in obese subjects. CONCLUSIONS: Obesity is associated with impaired aspirin responsiveness, largely because of body size. Impaired inhibition of platelet activation by conventional low-dose aspirin may affect antithrombotic efficacy.


Subject(s)
Aspirin/administration & dosage , Obesity/blood , Obesity/drug therapy , Platelet Activation/drug effects , Adult , Aspirin/pharmacokinetics , Aspirin/pharmacology , Biological Availability , Biomarkers/blood , Body Mass Index , Body Weight , Case-Control Studies , Computer Simulation , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Models, Biological , Obesity/pathology , Pilot Projects , Proof of Concept Study , Thromboxane A2/biosynthesis , Thromboxane B2/blood
8.
Biomed Khim ; 65(1): 63-66, 2019 Jan.
Article in Russian | MEDLINE | ID: mdl-30816099

ABSTRACT

Biosensor experiments on investigation of interaction between prostacyclin synthase (PGIS) and different proteins of the cytochrome P450 monooxygenase systems were perfomed. Interaction of PGIS with microsomal (CYP21A2, CYP2E1) and mitochondrial (CYP27A1, CYP11B1, CYP11B2, CYP11A1) cytochrome P450s was detected. Kinetic and equilibrium parameters of protein complexes formation were determined. Data obtained suggest an essential role of these hemoproteins interaction in regulation of prostacyclin and thromboxane A2 biosynthesis.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Intramolecular Oxidoreductases/metabolism , Humans , Microsomes/enzymology , Mitochondria/enzymology , Prostaglandins I/biosynthesis , Thromboxane A2/biosynthesis
9.
FASEB J ; 33(5): 6584-6595, 2019 05.
Article in English | MEDLINE | ID: mdl-30794438

ABSTRACT

LPS has been shown to elicit neuroinflammation associated with the up-regulation of the eicosanoid pathway in animal models; however, the regulatory mechanisms of TLR4 in brain neuroinflammatory conditions remain elusive. ß-Arrestins are key regulators of the GPCR signaling pathway and are involved in the leukotriene B4-induced leukocyte migration to initiate inflammatory response. However, the roles of ß-arrestins in eicosanoid regulation and related diseases are not clear. To address this issue, we conducted a study to investigate the effect of TLR4 on the eicosanoid pathway in ischemic stroke brain and to explore the underlying molecular regulation mechanism. Cerebral ischemia was produced by occlusion of the middle cerebral artery, followed by reperfusion for 24 h. We demonstrated that knockout of TLR4 improves ischemic stroke brain associated with eicosanoid down-regulation. Interestingly, genetic disruption of ß-arrestin-2 failed to decrease neuroinflammation in the damaged brain of TLR4-/- mice, which indicates the requirement of ß-arrestin-2 for TLR4 knockdown protection. Further study showed that the negative regulation of phosphorylated (phospho-)ERK1/2 and phospho-cytosolic phospholipase A2 α (cPLA2α) by TLR4 deficiency was eliminated by genetic disruption of ß-arrestin-2. In addition, ß-arrestin-2 deficiency reversed the reduction of colocalization of phospho-ERK1/2 with phospho-cPLA2α in TLR4-/- mice following ischemic stroke. Mechanistic studies indicated that ß-arrestin-2 specifically colocalized and associated with ERK1/2 to prevent ERK1/2-dependent cPLA2α activation following ischemic injury, and ß-arrestin-2 deficiency blocked the negative regulation of phospho-ERK1/2, revived the association of phospho-ERK1/2 with phospho-cPLA2α, and subsequently increased the prostaglandin E2 and thromboxane A2 production remarkably. Our findings may provide novel insights that ß-arrestin-2 is responsible for ischemic brain improvement in TLR4-/- mice via negative regulation of eicosanoid production.-Xiang, Y., Wei, X., Du, P., Zhao, H., Liu, A., Chen, Y. ß-Arrestin-2-ERK1/2 cPLA2α axis mediates TLR4 signaling to influence eicosanoid induction in ischemic brain.


Subject(s)
Brain Ischemia/metabolism , Dinoprostone/biosynthesis , Group IV Phospholipases A2/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Thromboxane A2/biosynthesis , Toll-Like Receptor 4/metabolism , beta-Arrestin 2/metabolism , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Dinoprostone/genetics , Group IV Phospholipases A2/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Stroke/genetics , Stroke/metabolism , Stroke/pathology , Thromboxane A2/genetics , Toll-Like Receptor 4/genetics , beta-Arrestin 2/genetics
10.
Eur J Pharmacol ; 852: 1-13, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30797789

ABSTRACT

Aspirin eugenol ester (AEE) was a promising drug candidate for treating inflammation, pain and fever and preventing cardiovascular diseases with fewer side effects than its precursors. Previous researches indicated that AEE could markedly inhibit agonist-induced platelet aggregation in vitro and ex vivo, however, the anti-platelet aggregation mechanisms of AEE remain to be defined. Here, AEE in vitro effects on agonist-induced granule-secretion, intercellular Ca2+ mobilization and thromboxane A2 (TXA2) generation were examined. Vasodilator-stimulated phosphoprotein (VASP), mitogen-activated protein kinase (MAPK), Akt, Sirt 1 and CD40L expressions were also studied. In agonist-activated platelets in vitro, AEE markedly attenuated granule secretion markers (P-selectin expression and ATP release), intercellular Ca2+ mobilization and thromboxane B2 (TXB2) formation. AEE also attenuated CD40L activation, suppressed extracellular-signal-regulated protein kinase 2 (ERK2), c-Jun N-terminal kinase 1 (JNK1) and Akt phosphorylation, and recovered Sirt1 expression, but the activation of p38, VASPSer157 and VASPSer239, and the levels of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) were not affected by AEE. Overall, this study demonstrates that AEE inhibits agonist-induced platelet aggregation in vitro by regulating PI3K/Akt, MAPK and Sirt 1/CD40L pathways.


Subject(s)
Aspirin/analogs & derivatives , CD40 Ligand/metabolism , Eugenol/analogs & derivatives , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , Animals , Aspirin/pharmacology , Calcium/metabolism , Caspase 3/metabolism , Cell Adhesion Molecules/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Eugenol/pharmacology , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar , Thromboxane A2/biosynthesis
11.
Am J Cardiovasc Drugs ; 18(6): 457-472, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29987548

ABSTRACT

Patients surviving an acute coronary syndrome (ACS) remain at increased risk of ischemic events long term. This paper reviews current evidence and guidelines for oral antiplatelet therapy for secondary prevention following ACS, with respect to decreased risk of ischemic events versus bleeding risk according to individual patient characteristics and risk factors. Specifically, data are reviewed from clinical studies of clopidogrel, prasugrel, ticagrelor and vorapaxar, as well as the results of systematic reviews and meta-analyses looking at the benefits and risks of oral antiplatelet therapy, and the relative merits of shorter versus longer duration of dual antiplatelet therapy, in different patient groups.


Subject(s)
Acute Coronary Syndrome/prevention & control , Platelet Aggregation Inhibitors/administration & dosage , Secondary Prevention/methods , Acute Coronary Syndrome/epidemiology , Adenosine Diphosphate/metabolism , Administration, Oral , Aging , Blood Platelets/metabolism , Cyclooxygenase 1/biosynthesis , Diabetes Mellitus/epidemiology , Drug Administration Schedule , Hemorrhage/chemically induced , Humans , Platelet Aggregation Inhibitors/adverse effects , Practice Guidelines as Topic , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/adverse effects , Receptor, PAR-1/biosynthesis , Receptors, Purinergic P2Y12/biosynthesis , Renal Insufficiency/epidemiology , Risk Factors , Thrombin/metabolism , Thromboxane A2/biosynthesis , Time Factors , Vascular Diseases/epidemiology
12.
Int J Mol Med ; 42(1): 615-624, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693158

ABSTRACT

Platelet activation and subsequent accumulation at sites of vascular injury are central to thrombus formation, which is considered to be a trigger of several cardiovascular diseases. Callicarpa nudiflora (C. nudiflora) Hook is a traditional Chinese medicinal herb for promoting blood circulation by removing blood stasis. In our previous study, several compounds extracted from this herb, including luteolin­4'­O­ß­D­glucopyranoside (LGP), were revealed to exert inhibitory effects on adenosine diphosphate (ADP)­induced platelet aggregation. The aim of present study was to confirm these antiplatelet effects and elucidate the potential mechanisms. Using a platelet­aggregation assay, it was revealed that LGP significantly inhibited platelet aggregation induced by ADP, U46619 and arachidonic acid. It was also found that LGP exhibited marked inhibitory effects on the activation of αIIbß3 integrin, the secretion of serotonin from granules, and the synthesis of thromboxane A2. In addition, the results showed that LGP suppressed Ras homolog family member A and phosphoinositide 3­kinase/Akt/glycogen synthase kinase 3ß signal transduction. Data from a radiolabeled ligand­binding assay indicated that LGP exhibited apparent competing effects on thromboxane receptor (TP) and P2Y12 receptors. In conclusion, the data presented here demonstrated that LGP, a natural compound from C. nudiflora Hook, inhibited the development of platelet aggregation and amplification of platelet activation. These inhibitory effects may be associated with its dual­receptor inhibition on P2Y12 and TP receptors.


Subject(s)
Glucosides/pharmacology , Luteolin/pharmacology , Platelet Activation/drug effects , Receptors, Purinergic P2Y12/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Arachidonic Acid/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Glucosides/chemistry , Glycogen Synthase Kinase 3 beta/metabolism , Hydrazines/metabolism , Luteolin/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Serotonin/metabolism , Signal Transduction/drug effects , Thromboxane A2/biosynthesis , Tritium , rhoA GTP-Binding Protein/metabolism
13.
Sci Rep ; 8(1): 3494, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472624

ABSTRACT

Citalopram prevents serotonin (5-HT) uptake into platelets by blocking the serotonin reuptake transporter (SERT). Although some clinical data suggest that selective serotonin reuptake inhibitors (SSRIs) may affect haemostasis and thrombosis, these poorly-characterised effects are not well understood mechanistically and useful in vitro data is limited. We sought to determine whether the inhibitory effects of citalopram on platelets are mediated via its pharmacological inhibition of 5-HT transport. We quantified the inhibitory potency of (RS)-, (R)- and (S)-citalopram on platelet function. If SERT blockade is the primary mechanism for citalopram-mediated platelet inhibition, these potencies should show quantitative congruence with inhibition of 5-HT uptake. Our data show that citalopram inhibits platelet aggregation, adhesion and thromboxane production with no difference in potency between (R)- and (S)-isomers. By contrast, citalopram had a eudysmic ratio of approximately 17 (S > R) for SERT blockade. Furthermore, nanomolar concentrations of citalopram inhibited 5-HT uptake into platelets but had no effect on other platelet functions, which were inhibited by micromolar concentrations. Our data indicate that citalopram-induced inhibition of platelets in vitro is not mediated by blockade of 5-HT transport. This raises a new question for future investigation: by what mechanism(s) does citalopram inhibit platelets?


Subject(s)
Citalopram/pharmacology , Platelet Aggregation/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin/genetics , Animals , Blood Platelets/drug effects , Blood Platelets/pathology , Healthy Volunteers , Humans , Mice , Phosphorylation , Platelet Aggregation/genetics , Rabbits , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/drug effects , Selective Serotonin Reuptake Inhibitors/antagonists & inhibitors , Signal Transduction/drug effects , Thromboxane A2/biosynthesis , Thromboxane A2/genetics
14.
Eur J Pharmacol ; 814: 87-94, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28780058

ABSTRACT

In this work, we hypothesized that cyclooxygenase (COX) activity can be regulated by nitric oxide (NO) and hydrogen peroxide (H2O2). In the renal hypertension (2K-1C), phenylephrine (PE)-induced contraction was lower than in normotensive (2K) rat aortas. This impaired contraction is due to NO/H2O2- induced vasodilation. We evaluated the effects of H2O2 on the activity of COX and endothelial NO-Synthase (eNOS) in 2K-1C rat aortas stimulated with PE. Responses for PE or H2O2 were evaluated in 2K-1C and 2K rat aortas, without or with inhibitors for COX (Indomethacin) or eNOS (L-NAME). COX isoforms expression was evaluated by Western blotting. eNOS inhibition was tested on thromboxane A2 (TXA2) and prostacyclin (PGI2) production. PE-induced contraction was lower in 2K-1C than in 2K. Indomethacin reduced PE-induced contraction in 2K, but it had no effect in 2K-1C. L-NAME reversed indomethacin-induced effect in 2K and it normalized PE-induced contraction in 2K-1C to the normotensive levels. COX-1 and COX-2 expression, TXA2 and PGI2 production were higher in 2K-1C than in 2K. eNOS inhibition did no modify TXA2/PGI2 production. In low concentrations, H2O2 induced relaxation only in 2K that was abolished by L-NAME while the contractions induced by high concentrations were abolished by indomethacin in both 2K and 2K-1C. The activity/expression of COX, and TXA2/PGI2 production were increased in 2K-1C, which were not modified by eNOS. High levels of H2O2 increased the endothelial COX activity, which induced contraction. Therefore, an high increase in H2O2 production may increase COX-induced vasoconstriction rather than eNOS-induced relaxation, which might contribute to aggravate hypertension.


Subject(s)
Aorta/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Hydrogen Peroxide/pharmacology , Hypertension, Renal/metabolism , Nitric Oxide Synthase Type III/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Aorta/metabolism , Aorta/physiopathology , Blood Pressure/drug effects , Cyclooxygenase Inhibitors/pharmacology , Enzyme Activation/drug effects , Epoprostenol/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Hypertension, Renal/physiopathology , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Rats , Thromboxane A2/biosynthesis , Vasoconstriction/drug effects , Vasodilation/drug effects
15.
BMC Complement Altern Med ; 17(1): 396, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28797264

ABSTRACT

BACKGROUND: Arachidonic acid metabolism by cyclooxygenase (COX) is a major pathway for blood platelets' activation, which is associated with pro-thrombotic platelet activity and the production of pro-inflammatory mediators. Inhibition of COX activity is one of the major means of anti-platelet pharmacotherapy preventing arterial thrombosis and reducing the incidence of cardiovascular events. Recent studies have presented that a silymarin (standardized extract of Milk thistle (Silybum marianum)) can inhibit the COX pathway. Accordingly, the aim of our study was to determine the effects of three major flavonolignans (silybin, silychristin and silydianin) on COX pathway activity in blood platelets. METHODS: We determined the effect of flavonolignans on arachidonic acid induced blood platelet aggregation, COX pathway metabolites formation, as well as COX activity in platelets. Additionally, we analysed the potential mechanism of this interaction using the bioinformatic ligand docking method. RESULTS: We observed that tested compounds decrease the platelet aggregation level, both thromboxane A2 and malondialdehyde formation, as well as inhibit the COX activity. The strongest effect was observed for silychristin and silybin. In our in silico study we showed that silychristin and silybin have conformations which interact with the active COX site as competitive inhibitors, blocking the possibility of substrate binding. CONCLUSIONS: The results obtained from this study clearly present the potential of flavonolignans as novel antiplatelet and anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonic Acid/blood , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Silybum marianum/chemistry , Silymarin/pharmacology , Binding, Competitive , Blood Platelets/metabolism , Flavonolignans/pharmacology , Humans , Inflammation Mediators/blood , Malondialdehyde/blood , Molecular Docking Simulation , Plant Extracts/pharmacology , Prostaglandin-Endoperoxide Synthases/blood , Silybin , Silymarin/chemistry , Thromboxane A2/biosynthesis
16.
J Infect ; 74(6): 599-608, 2017 06.
Article in English | MEDLINE | ID: mdl-28267572

ABSTRACT

OBJECTIVES: Platelets orchestrate the inflammatory activities of neutrophils, possibly contributing to pulmonary and myocardial damage during severe pneumococcal infection. This study tested the hypothesis that the pneumococcal toxin, pneumolysin (Ply), activates production of platelet-activating factor (PAF) and thromboxane A2 (TxA2) by neutrophils, these bioactive lipids being potential mediators of neutrophil:platelet (NP) networking. METHODS: The effects of recombinant Ply (10-80 ng mL-1) on the production of PAF and TxA2 by isolated neutrophils were measured using ELISA procedures, and NP aggregation by flow cytometry. RESULTS: Exposure of neutrophils to Ply induced production of PAF and, to a lesser extent, TxA2, achieving statistical significance at ≥20 ng mL-1 of the toxin. In the case of NP interactions, Ply promoted heterotypic aggregation which was dependent on upregulation of P-selectin (CD62P) and activation of protease-activated receptor 1 (PAR1), attaining statistical significance at ≥10 ng mL-1 of the toxin, but did not involve either PAF or TxA2. CONCLUSION: Ply induces synthesis of PAF and TxA2, by human neutrophils, neither of which appears to contribute to the formation of NP heterotypic aggregates in vitro, a process which is seemingly dependent on CD62P and PAR1. These pro-inflammatory activities of Ply may contribute to the pathogenesis of pulmonary and myocardial injury during severe pneumococcal infection.


Subject(s)
Blood Platelets/physiology , Cell Aggregation , Neutrophils/physiology , Platelet Aggregation , Streptolysins/pharmacology , Streptolysins/physiology , Bacterial Proteins/pharmacology , Bacterial Proteins/physiology , Carrier Proteins/biosynthesis , Cell Survival , DNA-Binding Proteins , Humans , Neutrophil Activation , P-Selectin/genetics , Platelet Activation , Recombinant Proteins/pharmacology , Streptococcus pneumoniae/chemistry , Thromboxane A2/biosynthesis
17.
J Cell Mol Med ; 21(8): 1584-1592, 2017 08.
Article in English | MEDLINE | ID: mdl-28244682

ABSTRACT

Extracellular histones are mediators of inflammation, tissue injury and organ dysfunction. Interactions between circulating histones and vascular endothelial cells are key events in histone-mediated pathologies. Our aim was to investigate the implication of extracellular histones in the production of the major vasoactive compounds released by human endothelial cells (HUVECs), prostanoids and nitric oxide (NO). HUVEC exposed to increasing concentrations of histones (0.001 to 100 µg/ml) for 4 hrs induced prostacyclin (PGI2) production in a dose-dependent manner and decreased thromboxane A2 (TXA2) release at 100 µg/ml. Extracellular histones raised cyclooxygenase-2 (COX-2) and prostacyclin synthase (PGIS) mRNA and protein expression, decreased COX-1 mRNA levels and did not change thromboxane A2 synthase (TXAS) expression. Moreover, extracellular histones decreased both, eNOS expression and NO production in HUVEC. The impaired NO production was related to COX-2 activity and superoxide production since was reversed after celecoxib (10 µmol/l) and tempol (100 µmol/l) treatments, respectively. In conclusion, our findings suggest that extracellular histones stimulate the release of endothelial-dependent mediators through an up-regulation in COX-2-PGIS-PGI2 pathway which involves a COX-2-dependent superoxide production that decreases the activity of eNOS and the NO production. These effects may contribute to the endothelial cell dysfunction observed in histone-mediated pathologies.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Epoprostenol/agonists , Histones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Thromboxane A2/antagonists & inhibitors , Celecoxib/pharmacology , Cyclic N-Oxides/pharmacology , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Epoprostenol/biosynthesis , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/genetics , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Spin Labels , Superoxides/antagonists & inhibitors , Superoxides/metabolism , Thromboxane A2/biosynthesis , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/metabolism
18.
Blood ; 129(9): 1197-1209, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28028021

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are expressed in platelets and are activated downstream of physiological agonists. Pharmacological and genetic evidence indicate that MAPKs play a significant role in hemostasis and thrombosis, but it is not well understood how MAPKs are activated upon platelet stimulation. Here, we show that apoptosis signal-regulating kinase 1 (ASK1), a member of the MAP3K family, is expressed in both human and murine platelets. ASK1 is rapidly and robustly activated upon platelet stimulation by physiological agonists. Disruption of Ask1 (Ask1-/- ) resulted in a marked functional defect in platelets. Ask1-/- platelets showed an impaired agonist-induced integrin αIIbß3 activation and platelet aggregation. Although there was no difference in Ca2+ rise, platelet granule secretion and thromboxane A2 (TxA2) generation were significantly attenuated in Ask1-/- platelets. The defective granule secretion observed in Ask1-/- platelets was a consequence of impaired TxA2 generation. Biochemical studies showed that platelet agonists failed to activate p38 MAPK in Ask1-/- platelets. On the contrary, activation of c-Jun N-terminal kinases and extracellular signal-regulated kinase 1/2 MAPKs was augmented in Ask1-/- platelets. The defect in p38 MAPK results in failed phosphorylation of cPLA2 in Ask1-/- platelets and impaired platelet aggregate formation under flow. The absence of Ask1 renders mice defective in hemostasis as assessed by prolonged tail-bleeding times. Deletion of Ask1 also reduces thrombosis as assessed by delayed vessel occlusion of carotid artery after FeCl3-induced injury and protects against collagen/epinephrine-induced pulmonary thromboembolism. These results suggest that the platelet Ask1 plays an important role in regulation of hemostasis and thrombosis.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Platelet Activation/physiology , Thromboxane A2/biosynthesis , Animals , Cytoplasmic Granules/metabolism , Female , Flow Cytometry , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Sci Rep ; 6: 38757, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934923

ABSTRACT

The thromboxane (Tx) A2 pathway is a major contributor to the amplification of initial platelet activation and is therefore a key drug target. To identify potent small-molecule inhibitors of the thromboxane prostaglandin (TP) receptor, we screened a small steroidal saponin library using U46619-induced rat platelet aggregation assays. Timosaponin AIII (TAIII) was identified as a potent inhibitor of U46619-induced rat platelet aggregation and exhibited superior selectivity for the TP receptor versus other G protein-coupled receptors and a PKC activator. TAIII inhibited U46619-induced rat platelet aggregation independent of increases in cAMP and cGMP and the inhibition of TxA2 production. Both PKC and PLC activators restored TAIII-inhibited platelet aggregation, whereas TAIII did not inhibit platelet aggregation induced by co-activation of the G12/13 and Gz pathways. Furthermore, TAIII did not affect the platelet shape change or ROCK2 phosphorylation evoked by low-dose U46619. In vivo, TAIII prolonged tail bleeding time, reduced the mortality of animals with acute pulmonary thromboembolism and significantly reduced venous thrombus weight. Our study suggests that TAIII, by preferentially targeting Gq-mediated PLC/PKC signaling from the TP receptor, induces stronger in vitro antiplatelet activity and in vivo antithrombotic effects and may be an excellent candidate for the treatment of thrombotic disorders.


Subject(s)
Antithrombins/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Platelet Aggregation Inhibitors/pharmacology , Receptors, Thromboxane A2, Prostaglandin H2/physiology , Saponins/pharmacology , Signal Transduction/physiology , Steroids/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Drug Synergism , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Thromboxane A2/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...